Language selection

Search

Patent 2633038 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2633038
(54) English Title: ANTI-MN ANTIBODIES AND METHODS OF USING SAME
(54) French Title: ANTICORPS ANTI-MN ET LEURS PROCEDES D'UTILISATION
Status: Deemed Abandoned and Beyond the Period of Reinstatement - Pending Response to Notice of Disregarded Communication
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07K 16/28 (2006.01)
  • A61K 39/395 (2006.01)
(72) Inventors :
  • TAMBURINI, PAUL (United States of America)
  • RANGES, GERALD (United States of America)
  • ADNANE, LILA (United States of America)
  • MCCABE, TIMOTHY (United States of America)
  • TRAIL, PAMELA (United States of America)
  • HA, SHA (United States of America)
(73) Owners :
  • BAYER HEALTHCARE LLC
(71) Applicants :
  • BAYER HEALTHCARE LLC (Germany)
(74) Agent: BORDEN LADNER GERVAIS LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2006-12-12
(87) Open to Public Inspection: 2007-06-21
Examination requested: 2008-07-28
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2006/047445
(87) International Publication Number: WO 2007070538
(85) National Entry: 2008-06-11

(30) Application Priority Data:
Application No. Country/Territory Date
60/749,716 (United States of America) 2005-12-12

Abstracts

English Abstract


The invention provides antibodies having an antigenic binding site
specifically directed against an MN protein, and
methods for using such antibodies in treating and diagnosing an MN-related
disorder.


French Abstract

La présente invention concerne des anticorps comprenant un site de liaison antigénique spécifiquement dirigé contre une protéine de MN. L'invention concerne également des procédés d'utilisation de tels anticorps pour le diagnostic et le traitement d'un trouble lié aux MN.

Claims

Note: Claims are shown in the official language in which they were submitted.


WHAT IS CLAIMED IS:
1. An antibody or antibody fragment having an antigenic binding site
specifically directed against an MN protein, wherein the antigenic binding
site includes at
least one CDR1, CDR2, or CDR3:
(a) said CDR1 is selected from the group consisting of SEQ ID NOS: 57, 58, 59,
60, 61, 62, 77, 80, 81, 86, 87, 88, 89, 98, 99, 104, 107, 108, and an amino
acid sequence
having greater than about 80% sequence identity to any of SEQ ID NOS: 57, 58,
59, 60,
61, 62, 77, 80, 81, 86, 87, 88, 89, 98, 99, 104, 107, or 108;
(b) said CDR2 is selected from the group consisting of SEQ ID NOS: 63, 64, 65,
66, 67, 68, 69, 78, 82, 83, 90, 91, 92, 93, 100, 101, 105, 109, 110 and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
63, 64,
65, 66, 67, 68, 69, 78, 82, 83, 90, 91, 92, 93, 100, 101, 105, 109, or 110;
and
(c) said CDR3 is selected from the group consisting of SEQ ID NOS: 70, 71, 72,
73, 74, 75, 76, 79, 84, 85, 94, 95, 96, 97, 102, 103, 106, 111, 112 and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
70, 71,
72, 73, 74, 75, 76, 79, 84, 85, 94, 95, 96, 97, 102, 103, 106, 111, or 112.
2. The antibody or antibody fragment according to claim 1, wherein the
antigenic binding site comprises a heavy chain variable region CDR selected
from the
group consisting of: SEQ ID NOS: 57-85 and an amino acid sequence having
greater than
about 80% sequence identity to any of SEQ ID NOS: 57-85.
3. The antibody or antibody fragment according to claim 1, wherein the
antigenic binding site comprises a light chain variable region CDR selected
from the group
99

consisting of: SEQ ID NOS: 86-112 and an amino acid sequence having greater
than about
80% sequence identity to any of SEQ ID NOS: 86-112.
4. The antibody or antibody fragment according to claim 1, wherein the
antigenic binding site comprises a set of CDR sequences selected from -the
group
consisting of:
(a) (a) [3ee9] SEQ ID NOS: 57, 63, 70, 89, 93, and 97;
(b) [3ef2] SEQ ID NOS: 58, 64, 71, 107, 109, and 111;
(c) [1e4] SEQ ID NOS: 59, 65, 72, 107, 109, and 111;
(d) [3a4] SEQ ID NOS: 60, 66, 73, 108, 110, and 112;
(e) [3ab4] SEQ ID NOS: 61, 67, 74, 87, 91, and 95;
(f) [3ah10] SEQ ID NOS: 61, 68, 75, 88, 92, and 96;
(g) [3bb2] SEQ ID NOS: 62, 69, 76, 98, 100, and 102;
(h) [1aa1] SEQ ID NOS: 77, 78, 79, 86, 90, and 94;
(i) [5a6] SEQ ID NOS: 80, 82, 84, 99, 101, and 103; and
(j) [5aa3] SEQ ID NOS: 81, 83, 85, 104, 105, and 106.
5. The antibody or antibody fragment according to claim 1, wherein the
antigenic binding site comprises a set of heavy chain CDR sequences selected
from the
group consisting of:
(a) [3ee9] SEQ ID NOS: 57, 63, and 70;
(b) [3ef2] SEQ ID NOS: 58, 64, and 71;
(c) [1e4] SEQ ID NOS: 59, 65, and 72;
(d) [3a4] SEQ ID NOS: 60, 66, and 73;
(e) [3ab4] SEQ ID NOS: 61, 67, and 74;
100

(f) [3ah 10] SEQ ID NOS: 61, 68, and 75;
(g) [3bb2] SEQ ID NOS: 62, 69, and 76;
(h) [1aa1] SEQ ID NOS: 77, 78, and 79;
(i) [5a6] SEQ ID NOS: 80, 82, and 84; and
(j) [5aa3] SEQ ID NOS: 81, 83, and 85.
6. The antibody or antibody fragment according to claim 1, wherein the
antigenic binding site comprises a set of light chain CDR sequences selected
from the
group consisting of:
(a) [3ee9] SEQ ID NOS: 89, 93, and 97;
(b) [3ef2] SEQ ID NOS: 107, 109, and 111;
(c) [1e4] SEQ ID NOS: 107, 109, and 111;
(d) [3a4] SEQ ID NOS: 108, 110, and 112;
(e) [3ab4] SEQ ID NOS: 87, 91, and 95;
(f) [3ah10] SEQ ID NOS: 88, 92, and 96;
(g) [3bb2] SEQ ID NOS: 98, 100, and 102;
(h) [1aa1] SEQ ID NOS: 86, 90, and 94;
(i) [5a6] SEQ ID NOS: 99, 101, and 103; and
(j) [5aa3] SEQ ID NOS: 104, 105, and 106.
7. The antibody or antibody fragment according to claim 1, wherein the
antibody or antibody fragment binds to the MN protein with a dissociation
constant of
about 0.15 nM to about 50 nM.
101

8. The antibody or antibody fragment according to claim 1, wherein the
antibody is an IgG.
9. The antibody or antibody fragment according to claim 1, wherein the
antibody is an-IgG1, IgG2a, IgG2b, IgG3, IgM, IgD, IgE, IgA, IgM Fab fragment,
F(ab')2
fragment, scFv fragment, Fv fragment, a diabody, linear antibody, single-chain
antibody,
biospecific antibody, chimeric antibody, or multispecific antibody.
10. The antibody or antibody fragment according to claim 1, wherein the
antibody or antibody fragment is humanized.
11. The antibody or antibody fragment according to claim 1, wherein the
CDR1, CDR2, and CDR3 are non-human.
12. A composition comprising an antibody or antibody fragment thereof
according to claim 1 and one or more pharmaceutical auxiliary substance.
13. The antibody or antibody fragment according to claim 1, wherein the
sequence identity is greater than about 85%.
14. The antibody or antibody fragment according to claim 1, wherein the
sequence identity is greater than about 90%.
15. The antibody or antibody fragment according to claim 1, wherein the
sequence identity is greater than about 95%.
102

16. The antibody or antibody fragment according to claim 1, wherein the
sequence identity is greater than about 99%.
17. A composition reactive against MN protein comprising an antibody or
antibody fragment having an antigenic binding site specifically directed
against an MN
protein conjugated to a cytotoxic agent , wherein the antigenic binding site
includes at
least one CDR1, CDR2, or CDR3:
(a) said CDR1 is selected from the group consisting of SEQ ID NOS: 57, 58,
59, 60, 61, 62, 77, 80, 81, 86, 87, 88, 89, 98, 99, 104, 107, 108, and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
57, 58,
59, 60, 61, 62, 77, 80, 81, 86, 87, 88, 89, 98, 99, 104, 107, or 108;
(b) said CDR2 is selected from the group consisting of SEQ ID NOS: 63, 64,
65, 66, 67, 68, 69, 78, 82, 83, 90, 91, 92, 93, 100, 101, 105, 109, 110 and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
63, 64,
65, 66, 67, 68, 69, 78, 82, 83, 90, 91, 92, 93, 100, 101, 105, 109, or 110;
and
(c) said CDR3 is selected from the group consisting of SEQ ID NOS: 70, 71,
72, 73, 74, 75, 76, 79, 84, 85, 94, 95, 96, 97, 102, 103, 106, 111, 112 and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
70, 71,
72, 73, 74, 75, 76, 79, 84, 85, 94, 95, 96, 97, 102, 103, 106, 111, or 112.
18. The composition according to claim 17, wherein the antigenic binding site
comprises a heavy chain variable region CDR selected from the group consisting
of: SEQ
ID NOS: 57-85 and an amino acid sequence having greater than about 80%
sequence
identity to any of SEQ ID NOS: 57-85.
103

19. The composition according to claim 17, wherein the antigenic binding site
comprises a light chain variable region CDR selected from the group consisting
of: SEQ
ID NOS: 86-112 and an amino acid sequence having greater than about 80%
sequence
identity to any of SEQ ID NOS: 86-112.
20. The composition according to claim 17, wherein the antigenic binding site
comprises a set of CDR sequences selected from the group consisting of:
(a) [3ee9] SEQ ID NOS: 57, 63, 70, 89, 93, and 97;
(b) [3ef2] SEQ ID NOS: 58, 64, 71, 107, 109, and 111;
(c) [1e4] SEQ ID NOS: 59, 65, 72, 107, 109, and 111;
(d) (d) [3a4] SEQ ID NOS: 60, 66, 73, 108, 110, and 112;
(e) [3ab4] SEQ ID NOS: 61, 67, 74, 87, 91, and 95;
(f) [3ah10] SEQ ID NOS: 61, 68, 75, 88, 92, and 96;
(g) [3bb2] SEQ ID NOS: 62, 69, 76, 98, 100, and 102;
(h) [1aa1] SEQ ID NOS: 77, 78, 79, 86, 90, and 94;
(i) [5a6] SEQ ID NOS: 80, 82, 84, 99, 101, and 103; and
(j) [5aa3] SEQ ID NOS: 81, 83, 85, 104, 105, and 106.
21. The antibody or antibody fragment according to claim 17, wherein the
antigenic binding site comprises a set of heavy chain CDR sequences selected
from the
group consisting of:
a) [3ee9] SEQ ID NOS: 57, 63, and 70;
b) [3ef2] SEQ ID NOS: 58, 64, and 71;
c) [1e4] SEQ ID NOS: 59, 65, and 72;
104

d) [3a4] SEQ ID NOS: 60, 66, and 73;
e) [3ab4] SEQ ID NOS: 61, 67, and 74;
f) [3ah 10] SEQ ID NOS: 61, 68, and 75;
g) [3bb2] SEQ ID NOS: 62, 69, and 76;
h) [1aa1] SEQ ID NOS: 77, 78, and 79;
i) [5a6] SEQ ID NOS: 80, 82, and 84; and
j) [5aa3] SEQ ID NOS: 81, 83, and 85.
22. The antibody or antibody fragment according to claim 17, wherein the
antigenic binding site comprises a set of light chain CDR sequences selected
from the
group consisting of:
a) [3ee9] SEQ ID NOS: 89, 93, and 97;
b) [3ef2] SEQ ID NOS: 107, 109, and 111;
c) [1e4] SEQ ID NOS: 107, 109, and 111;
d) [3a4] SEQ ID NOS: 108, 110, and 112;
e) [3ab4] SEQ ID NOS: 87, 91, and 95;
f) [3ah10] SEQ ID NOS: 88, 92, and 96;
g) [3bb2] SEQ ID NOS: 98, 100, and 102;
h) [1aa1] SEQ ID NOS: 86, 90, and 94;
i) [5a6] SEQ ID NOS: 99, 101, and 103; and
j) [5aa3] SEQ ID NOS: 104, 105, and 106.
23. The composition according to claim 17, wherein the cytotoxic agent is
selected from the group consisting of: monomethylauristatin-E or functional
analog
thereof, monomethylauristatin-F or functional analog thereof, aplidin,
azaribine,
105

anastrozole, azacytidine, bleomycin, bortezomib, bryostatin-1, busulfan,
calicheamycin,
camptothecin, 10-hydroxycamptothecin, carmustine, celebrex, chlorambucil,
cisplatin,
irinotecan (CPT-11), SN-38, carboplatin, cladribine, cyclophosphamide,
cytarabine,
dacarbazine, docetaxel, dactinomycin, daunomycin glucuronide, daunorubicin,
dexamethasone, diethylstilbestrol, doxorubicin, doxorubicin glucuronide,
epirubicin
glucuronide, ethinyl estradiol, estramustine, etoposide, etoposide
glucuronide, etoposide
phosphate, floxuridine (FUdR), 3',5'-O-dioleoyl-FudR (FUdR-dO), fludarabine,
flutamide,
fluorouracil, fluoxymesterone, gemcitabine, hydroxyprogesterone caproate,
hydroxyurea,
idarubicin, ifosfamide, L-asparaginase, leucovorin, lomustine,
mechlorethamine,
medroprogesterone acetate, megestrol acetate, melphalan, mercaptopurine, 6-
mercaptopurine, methotrexate, mitoxantrone, mithramycin, mitomycin, mitotane,
phenyl
butyrate, prednisone, procarbazine, paclitaxel, pentostatin, PSI-341,
semustine
streptozocin, tamoxifen, taxanes, taxol, testosterone propionate, thalidomide,
thioguanine,
thiotepa, teniposide, topotecan, uracil mustard, velcade, vinblastine,
vinorelbine,
vincristine, ricin, abrin, ribonuclease, onconase, rapLR1, DNase I,
Staphylococcal
enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria toxin,
Pseudomonas
exotoxin, Pseudomonas endotoxin, and combinations thereof.
24. The composition according to claim 17, wherein the cytotoxic agent is
monomethylauristatin-E or functional analog thereof.
25. The composition according to claim 17, wherein the antibody or antibody
fragment binds to the MN protein with a dissociation constant of about 0.15 nM
to about
50 nM.
106

26. The composition according to claim 17, wherein the antibody is an IgG.
27. The composition according to claim 17, wherein the antibody or antibody
fragment is an IgG1, IgG2a, IgG2b, IgG3, IgM, IgD, IgE, IgA, or IgM isotype,
an Fab
fragment, F(ab')2 fragment, scFv fragment, Fv fragment, a diabody, linear
antibody,
single-chain antibody, biospecific antibody, a chimeric antibody or
multispecific antibody.
28. The composition according to claim 17, wherein the antibody or antibody
fragment humanized.
29. The antibody or antibody fragment according to claim 17, wherein the
CDR1, CDR2, and CDR3 are non-human.
30. A composition for treating a subject having an MN-related cancer,
comprising an anti-cancer agent and an antibody or antibody fragment thereof
having an
antigenic binding site specifically directed against an MN protein conjugated
to a
cytotoxic agent, wherein the antigenic binding site includes at least one
CDR1, CDR2, or
CDR3:
a) said CDR1 is selected from the group consisting of SEQ ID NOS: 57, 58,
59, 60, 61, 62, 77, 80, 81, 86, 87, 88, 89, 98, 99, 104, 107, 108, and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
57, 58,
59, 60, 61, 62, 77, 80, 81, 86, 87, 88, 89, 98, 99, 104, 107, or 108;
b) said CDR2 is selected from the group consisting of SEQ ID NOS: 63, 64,
65, 66, 67, 68, 69, 78, 82, 83, 90, 91, 92, 93, 100, 101, 105, 109, 110 and an
amino acid
107

sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
63, 64,
65, 66, 67, 68, 69, 78, 82, 83, 90, 91, 92, 93, 100, 101, 105, 109, or 110;
and
c) said CDR3 is selected from the group consisting of SEQ ID NOS: 70, 71,
72, 73, 74, 75, 76, 79, 84, 85, 94, 95, 96, 97, 102, 103, 106, 111, 112 and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
70, 71,
72, 73, 74, 75, 76, 79, 84, 85, 94, 95, 96, 97, 102, 103, 106, 111, or 112.
31. The composition according to claim 30, wherein the antigenic binding site
comprises a heavy chain variable region CDR selected from the group consisting
of: SEQ
ID NOS: 57-85 and an amino acid sequence having greater than about 80%
sequence
identity to any of SEQ ID NOS: 57-85.
32. The composition according to claim 30, wherein the antigenic binding site
comprises a light chain variable region CDR selected from the group consisting
of: SEQ
ID NOS: 86-112 and an amino acid sequence having greater than about 80%
sequence
identity to any of SEQ ID NOS: 86-112.
33. The composition according to claim 30, wherein the antigenic binding site
comprises a set of CDR sequences selected from the group consisting of:
a) [3ee9] SEQ ID NOS: 57, 63, 70, 89, 93, and 97;
b) [3ef2] SEQ ID NOS: 58, 64, 71, 107, 109, and 111;
c) [1e4] SEQ ID NOS: 59, 65, 72, 107, 109, and 111;
d) [3a4] SEQ ID NOS: 60, 66, 73, 108, 110, and 112;
e) [3ab4] SEQ ID NOS: 61, 67, 74, 87, 91, and 95;
f) [3ah10) SEQ ID NOS: 61, 68, 75, 88, 92, and 96;
108

g) [3bb2] SEQ ID NOS: 62, 69, 76, 98, 100, and 102;
h) [1aa1] SEQ ID NOS: 77, 78, 79, 86, 90, and 94;
i) [5a6] SEQ ID NOS: 80, 82, 84, 99, 101, and 103; and
j) [5aa3] SEQ ID NOS: 81, 83, 85, 104, 105, and 106.
34. The composition according to claim 30, wherein the antigenic binding site
comprises a set of heavy chain CDR sequences selected from the group
consisting of:
a) [3ee9] SEQ ID NOS: 57, 63, and 70;
b) [3ef2] SEQ ID NOS: 58, 64, and 71;
c) [1e4] SEQ ID NOS: 59, 65, and 72;
d) [3a4] SEQ ID NOS: 60, 66, and 73;
e) [3ab4] SEQ ID NOS: 61, 67, and 74;
f) [3ah10] SEQ ID NOS: 61, 68, and 75;
g) [3bb2] SEQ ID NOS: 62, 69, and 76;
h) [1aa1] SEQ ID NOS: 77, 78, and 79;
i) [5a6] SEQ ID NOS: 80, 82, and 84; and
j) [5aa3] SEQ ID NOS: 81, 83, and 85.
35. The composition according to claim 30, wherein the antigenic binding site
comprises a set of light chain CDR sequences selected from the group
consisting of:
a) [3ee9] SEQ ID NOS: 89, 93, and 97;
b) [3ef2] SEQ ID NOS: 107, 109, and 111;
c) [1e4] SEQ ID NOS: 107, 109, and 111;
d) [3a4] SEQ ID NOS: 108, 110, and 112;
e) [3ab4] SEQ ID NOS: 87,91, and 95;
109

f) [3ah10] SEQ ID NOS: 88, 92, and 96;
g) [3bb2] SEQ ID NOS: 98, 100, and 102;
h) [1aa1] SEQ ID NOS: 86, 90, and 94;
i) [5a6] SEQ ID NOS: 99, 101, and 103; and
j) [5aa3] SEQ ID NOS: 104, 105, and 106.
36. The composition according to claim 30, wherein the anti-cancer agent is
selected from the group consisting of bleomycin, docetaxel (Taxotere),
doxorubicin,
edatrexate, eriotinib (Tarceva), etoposide, finasteride (Proscar), flutamide
(Eulexin),
gemcitabine (Gemzar), genitinib (Irresa), goserelin acetate (Zoladex),
granisetron (Kytril),
imatinib (Gleevec), irinotecan (Campto/Camptosar), ondansetron (Zofran),
paclitaxel
(Taxol), pegaspargase (Oncaspar), pilocarpine hydrochloride (Salagen),
porfimer sodium
(Photofrin), interleukin-2 (Proleukin), rituximab (Rituxan), topotecan
(Hycamtin),
trastuzumab (Herceptin), Triapine, vincristine, vinorelbine tartrate
(Navelbine), and
therapeutic antibodies or fragments thereof.
37. The composition according to claim 30, wherein the anti-cancer agent is an
anti-angiogenic agent selected from the group consisting of angiostatin,
bevacizumab
(Avastin ®), sorafenib (Nexavar ®), baculostatin, canstatin, maspin,
anti-VEGF antibodies
or peptides, anti-placental growth factor antibodies or peptides, anti-Flk-1
antibodies, anti-
Flt-1 antibodies or peptides, laminin peptides, fibronectin peptides,
plasminogen activator
inhibitors, tissue metalloproteinase inhibitors, interferons, interleukin 12,
IP-10, Gro-.beta.,
thrombospondin, 2-methoxyoestradiol, proliferin-related protein,
carboxiamidotriazole,
CM101, Marimastat, pentosan polysulphate, angiopoietin 2, interferon-alpha,
herbimycin
A, PNU145156E, 16K prolactin fragment, Linomide, thalidomide, pentoxifylline,
110

genistein, TNP-470, endostatin, paclitaxel, accutin, cidofovir, vincristine,
bleomycin,
AGM-1470, platelet factor 4 and minocycline.
38. The composition according to claim 30, wherein the anti-cancer agent is an
agent that blocks or inhibits a multi-drug resistance phenotype selected from
the group
consisting of tamoxifen, verapamil and cyclosporin A.
39. The composition according to claim 30, wherein the cytotoxic agent is
monomethylauristatin-E or functional analog thereof.
40. The composition according to any one of claims 31, wherein the cytotoxic
agent is monomethylauristatin-E or functional analog thereof.
41. The composition according to claim 30, wherein the antibody or antibody
fragment binds to the MN protein with a dissociation constant of about 0.15 nM
to about
50 nM.
42. The composition according to claim 30, wherein the antibody is an IgG.
43. The composition according to claim 30, wherein the antibody or antibody
fragment is an IgG1, IgG2a, IgG2b, IgG3, IgM, IgD, IgE, IgA, or IgM isotype, a
Fab
fragment, F(ab')2 fragment, scFv fragment, Fv fragment, a diabody, linear
antibody,
single-chain antibody, biospecific antibody, chimeric antibody or
multispecific antibody.
111

44. The composition according to claim 30, wherein the antibody or antibody
fragment is humanized.
45. The composition according to claim 30, wherein the CDR1, CDR2, and
CDR3 are non-human.
46. The composition according to claim 30, wherein the sequence identity is
greater than about 85%.
47. The composition according to claim 30, wherein the sequence identity is
greater than about 90%.
48. The composition according to claim 30, wherein the sequence identity is
greater than about 95%.
49. The composition according to claim 30, wherein the sequence identity is
greater than about 99%.
50. The composition according to claim 30, wherein the cancer is in the form
of a solid tumor.
51. The composition according to claim 50, wherein the solid tumor is in or
originating from the breast, respiratory tract, lung, brain, reproductive
organ, digestive
tract, colon, urinary tract, kidney, esophagus, cervix, eye, liver, skin,
head, neck, thyroid,
and parathyroid.
112

52. A method of diagnosing an MN-related disorder characterized by abnormal
MN levels comprising comparing the level of MN in a suspected diseased tissue
or cell
with the level of MN in a corresponding healthy tissue or cell, wherein an
abnormal MN
level in the suspected diseased tissue or cell is an indication of an MN-
related disorder,
said step of comparing further comprising detecting the level of MN in the
diseased tissue
and the healthy tissue with the antibody or antibody fragment according to
claim 1.
53. The method according to claim 52, wherein the MN-related disorder is a
cancer.
54. The method according to claim 52, wherein the cancer is in the form of a
solid tumor.
55. The method according to claim 52, wherein the solid tumor is in or
originating from the breast, respiratory tract, lung, brain, reproductive
organ, digestive
tract, colon, urinary tract, kidney, esophagus, cervix, eye, liver, skin,
head, neck, thyroid,
and parathyroid.
56. The method according to claim 52, wherein the step of comparing further
comprises the steps of:
a) detecting the level of MN protein in the healthy tissue;
b) detecting the level of MN protein in the suspected diseased tissue; and
c) comparing the levels of MN protein from (a) and (b),
113

wherein an elevated level of MN protein in the suspected diseased tissue as
compared to
the level of MN protein in the healthy tissue is indicative of the presence of
an MN-related
disorder.
57. The method according to claim 52, wherein the step of detecting is
achieved by a nuclear imaging modality.
58. The method according to claim 57, wherein the nuclear imaging modality is
SPECT or PET.
59. A kit comprising the antibody or antibody fragment according to claim 1,
and a set of instructions for using the kit in a method of treating an MN-
related disorder.
60. A kit comprising the composition according to claim 17 and a set of
instructions for using the kit in a method of treating an MN-related disorder.
61. A kit comprising the composition according to claim 30 and a set of
instructions for using the kit in a method of treating an MN-related disorder.
62. The composition according to claim 17, wherein the sequence identity is
greater than about 85%.
63. The composition according to claim 17, wherein the sequence identity is
greater than about 90%.
114

64. The composition according to claim 17, wherein the sequence identity is
greater than about 95%.
65. The composition according to claim 15, wherein the sequence identity is
greater than about 99%.
66. A method of treating an MN-related disorder characterized by an abnormal
level of MN in a subject comprising administering a therapeutically effective
amount of an
antibody or antibody fragment having an antigenic binding site specifically
directed
against an MN protein, wherein the antigenic binding site includes at least
one CDR1,
CDR2, or CDR3:
a) said CDR1 is selected from the group consisting of SEQ ID NOS: 57, 58,
59, 60, 61, 62, 77, 80, 81, 86, 87, 88, 89, 98, 99, 104, 107, 108, and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
57, 58,
59, 60, 61, 62, 77, 80, 81, 86, 87, 88, 89, 98, 99, 104, 107, or 108;
b) said CDR2 is selected from the group consisting of SEQ ID NOS: 63, 64,
65, 66, 67, 68, 69, 78, 82, 83, 90, 91, 92, 93, 100, 101, 105, 109, 110 and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
63, 64,
65, 66, 67, 68, 69, 78, 82, 83, 90, 91, 92, 93, 100, 101, 105, 109, or 110;
and
c) said CDR3 is selected from the group consisting of SEQ ID NOS: 70, 71,
72, 73, 74, 75, 76, 79, 84, 85, 94, 95, 96, 97, 102, 103, 106, 111, 112 and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
70, 71,
72, 73, 74, 75, 76, 79, 84, 85, 94, 95, 96, 97, 102, 103, 106, 111, or 112.
115

67. The method according to claim 66, wherein the antigenic binding site
comprises a heavy chain variable region CDR selected from the group consisting
of: SEQ
ID NOS: 57-85 and an amino acid sequence having greater than about 80%
sequence
identity to any of SEQ ID NOS: 57-85.
68. The method according to claim 66, wherein the antigenic binding site
comprises a light chain variable region CDR selected from the group consisting
of: SEQ
ID NOS: 86-112 and an amino acid sequence having greater than about 80%
sequence
identity to any of SEQ ID NOS: 86-112.
69. The method according to claim 66, wherein the antigenic binding site
comprises a set of CDR sequences selected from the group consisting of:
a) [3ee9] SEQ ID NOS: 57, 63, 70, 89, 93, and 97;
b) [3ef2] SEQ ID NOS: 58, 64, 71, 107, 109, and 111;
c) [1e4] SEQ ID NOS: 59, 65, 72, 107, 109, and 111;
d) [3a4] SEQ ID NOS: 60, 66, 73, 108, 110, and 112;
e) [3ab4] SEQ ID NOS: 61, 67, 74, 87, 91, and 95;
f) [3ah10] SEQ ID NOS: 61, 68, 75, 88, 92, and 96;
g) [3bb2] SEQ ID NOS: 62, 69, 76, 98, 100, and 102;
h) [1aa1] SEQ ID NOS: 77, 78, 79, 86, 90, and 94;
i) [5a6] SEQ ID NOS: 80, 82, 84, 99, 101, and 103; and
j) [5aa3] SEQ ID NOS: 81, 83, 85, 104, 105, and 106.
116

70. The antibody or antibody fragment according to claim 66, wherein the
antigenic binding site comprises a set of heavy chain CDR sequences selected
from the
group consisting of:
b) [3ee9] SEQ ID NOS: 57, 63, and 70;
c) [3ef2] SEQ ID NOS: 58, 64, and 71;
d) [1e4] SEQ ID NOS: 59, 65, and 72;
e) [3a4j SEQ ID NOS: 60, 66, and 73;
f) [3ab4] SEQ ID NOS: 61, 67, and 74;
g) [3ah10] SEQ ID NOS: 61, 68, and 75;
h) [3bb2] SEQ ID NOS: 62, 69, and 76;
i) [1aa1] SEQ ID NOS: 77, 78, and 79;
j) [5a6] SEQ ID NOS: 80, 82, and 84; and
k) [5aa3] SEQ ID NOS: 81, 83, and 85.
71. The antibody or antibody fragment according to claim 66, wherein the
antigenic binding site comprises a set of light chain CDR sequences selected
from the
group consisting of:
a) [3ee9] SEQ ID NOS: 89, 93, and 97;
b) [3ef2] SEQ ID NOS: 107, 109, and 111;
c) [le4] SEQ ID NOS: 107, 109, and 111;
d) [3a4] SEQ ID NOS: 108, 110, and 112;
e) [3ab4] SEQ ID NOS: 87, 91, and 95;
f) [3ah10] SEQ ID NOS: 88, 92, and 96;
g) [3bb2] SEQ ID NOS: 98, 100, and 102;
h) [1aa1] SEQ ID NOS: 86, 90, and 94;
117

i) [5a6] SEQ ID NOS: 99, 101, and 103; and
j) [5aa3] SEQ ID NOS: 104, 105, and 106.
72. The method according to claim 66, wherein the antibody or antibody
fragment binds to the MN protein with a dissociation constant of about 0.15 nM
to about
50 nM.
73. The method according to claim 66, wherein the antibody is an IgG.
74. The method according to claim 66, wherein the antibody or antibody
fragment is an IgG1, IgG2a, IgG2b, IgG3, IgM, IgD, IgE, IgA, or IgM, a Fab
fragment,
F(ab')2 fragment, scFv fragment, Fv fragment, a diabody, linear antibody,
single-chain
antibody, biospecific antibody, chimeric antibody or multispecific antibody.
75. The method according to claim 66, wherein the antibody or antibody
fragment is humanized.
76. The method according to claim 66, wherein the CDR1, CDR2, and CDR3
are non-human.
77. The method according to claim 66, wherein the sequence identity is greater
than about 85%.
78. The method according to claim 66, wherein the sequence identity is greater
than about 90%.
118

79. The method according to claim 66, wherein the sequence identity is greater
than about 95%.
80. The method according to claim 66, wherein the sequence identity is greater
than about 99%.
81. The method according to claim 66, wherein the antibody or antibody
fragment is conjugated to a cytotoxic agent.
82. The method according to claim 81, wherein the cytotoxic agent is selected
from the group consisting of: monomethylauristatin-E or functional analog
thereof, is
selected from the group consisting of: monomethylauristatin-E or functional
analog
thereof, monomethylauristatin-F or functional analog thereof, aplidin,
azaribine,
anastrozole, azacytidine, bleomycin, bortezomib, bryostatin-1, busulfan,
calicheamycin,
camptothecin, 10-hydroxycamptothecin, carmustine, celebrex, chlorambucil,
cisplatin,
irinotecan (CPT-1 1), SN-38, carboplatin, cladribine, cyclophosphamide,
cytarabine,
dacarbazine, docetaxel, dactinomycin, daunomycin glucuronide, daunorubicin,
dexamethasone, diethylstilbestrol, doxorubicin, doxorubicin glucuronide,
epirubicin
glucuronide, ethinyl estradiol, estramustine, etoposide, etoposide
glucuronide, etoposide
phosphate, floxuridine (FUdR), 3',5'-O-dioleoyl-FudR (FUdR-dO), fludarabine,
flutamide,
fluorouracil, fluoxymesterone, gemcitabine, hydroxyprogesterone caproate,
hydroxyurea,
idarubicin, ifosfamide, L-asparaginase, leucovorin, lomustine,
mechlorethamine,
medroprogesterone acetate, megestrol acetate, melphalan, mercaptopurine, 6-
mercaptopurine, methotrexate, mitoxantrone, mithramycin, mitomycin, mitotane,
phenyl
119

butyrate, prednisone, procarbazine, paclitaxel, pentostatin, PSI-341,
semustine
streptozocin, tamoxifen, taxanes, taxol, testosterone propionate, thalidomide,
thioguanine,
thiotepa, teniposide, topotecan, uracil mustard, velcade, vinblastine,
vinorelbine,
vincristine, ricin, abrin, ribonuclease, onconase, rapLR1, DNase I,
Staphylococcal
enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria toxin,
Pseudomonas
exotoxin, Pseudomonas endotoxin, and combinations thereof.
83. The method according to claim 82, wherein the cytotoxic agent is
monomethylauristatin-E or functional analog thereof.
84. The method according to claim 66, further comprising co-administering a-
cancer agent.
85. The method according to claim 84, wherein the anti-cancer agent is
selected from the group consisting of bleomycin, docetaxel (Taxotere),
doxorubicin,
edatrexate, erlotinib (Tarceva), etoposide, finasteride (Proscar), flutamide
(Eulexin),
gemcitabine (Gemzar), genitinib (Irresa), goserelin acetate (Zoladex),
granisetron (Kytril),
imatinib (Gleevec), irinotecan (Campto/Camptosar), ondansetron (Zofran),
paclitaxel
(Taxol), pegaspargase (Oncaspar), pilocarpine hydrochloride (Salagen),
porfimer sodium
(Photofrin), interleukin-2 (Proleukin), rituximab (Rituxan), topotecan
(Hycamtin),
trastuzumab (Herceptin), Triapine, vincristine, vinorelbine tartrate
(Navelbine), and
therapeutic antibodies or fragments thereof.
120

86. The method according to claim 84, wherein the anti-cancer agent is an
agent that blocks or inhibits a multi-drug resistance phenotype selected from
the group
consisting of tamoxifen, verapamil and cyclosporin A.
87. The method according to claim 66, wherein the MN-related disorder is a
cancer.
88. The method according to claim 87, wherein the cancer is a solid tumor
cancer.
89. The composition according to claim 88, wherein the solid tumor is in or
originating from the breast, respiratory tract, lung, brain, reproductive
organ, digestive
tract, colon, urinary tract, kidney, esophagus, cervix, eye, liver, skin,
head, neck, thyroid,
and parathyroid.
90. The antibody or antibody fragment according to claim 1, wherein the
antigenic binding site a pair of heavy chain variable and light chain variable
regions
selected from the group consisting of:
(a) the heavy chain variable region of SEQ ID NO: 133 and the light chain
variable region of SEQ ID NO: 134;
(b) the heavy chain variable region of SEQ ID NO: 135 and the light chain
variable region of SEQ ID NO:136;
(c) the heavy chain variable region of SEQ ID NO: 137 and the light chain
variable region of SEQ ID NO:138;
(d) the heavy chain variable region of SEQ ID NO: 139 and the light chain
121

variable region of SEQ ID NO:140;
(e) the heavy chain variable region of SEQ ID NO:141 and the light chain
variable region of SEQ ID NO: 142;
(f) the heavy chain variable region of SEQ ID NO: 143 and the light chain
variable region of SEQ ID NO:144;
(g) the heavy chain variable region of SEQ ID NO: 145 and the light chain
variable region of SEQ ID NO: 146;
(h) the heavy chain variable region of SEQ ID NO: 147 and the light chain
variable region of SEQ ID NO: 148;
(i) the heavy chain variable region of SEQ ID NO: 149 and the light chain
variable region of SEQ ID NO: 150; and
(j) the heavy chain variable region of SEQ ID NO: 151 and the light chain
variable region of SEQ ID NO:152.
91. An anti-MN IgG antibody encoded by the nucleotide sequence of SEQ ID
NO: 153.
122

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
TITLE OF THE INVENTION
ANTI-MN ANTIBODIBS AND METHODS OF US]NG SAME
INCORPORATION BY REFERENCE
This application claims the benefit of U.S. Provisional Patent Application No.
60/749,716 filed December 12, 2005.
The foregoing application, and all documents cited therein and all documents
cited
or referenced therein, and all documents cited or referenced herein ("herein
cited
documents"), and all documents cited or referenced in herein cited documents,
together
with any manufacturer's instructions, descriptions, product specifications,
and product
] 0 sheets for any products mentioned herein or in any document incorporated
by reference
herein, are hereby incorporated herein by reference, and may be employed in
the practice
of the invention.
BACKGROUND OF THE INVENTION
1. Field of the Invention
The present invention relates to antibodies and/or fragments thereof having
specificity for MN protein. The invention further relates to antibody and/or
immunoconjugate compositions and their use in treating, preventing, and/or
diagnosing
MN-related disorders, e.g. cancer.
2. Background
The occurrence of cancer is most commonly associated with aging whereby 65%
of all new cases of cancer are recorded for patients aged 65 and over. Cancer
is the second
leading cause of death in the United States, exceeded only by heart disease.
Indeed, the
American Cancer Society has estimated that 1 in 4 people will die from cancer
in the U.S.,
assuming current mortality rates remain static. In the U.S. alone, 1,399,790
new cases and
564,830 deaths from cancer are expected in 2006. The majority of these new
cases are
1

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
expected to be cancers of the colon (106,680), lung (172,570) and breast
(214,640).
Moreover, both the incidence and prevalence of cancer is predicted to increase
approximately 15% over the next ten years, reflecting an average growth rate
of 1.4%
(American Cancer Society, 2006).
One recently identified tumor associated antigen, MN, a cell surface protein,
has
been found to be expressed in a number of clinical carcinomas. For example, MN
has
been found to be ectopically expressed in 100% of renal cell carcinomas (Liao,
SY,
Cancer Res., 1997, 57:2827-2831), 100% of carcinomas of the esophagus (Turner
JR,
Hum. Pathol., 199, 28:740-744), greater than 90% of cervical carcinomas (Liao,
SY,
Cancer Res., 1997, 57:2827-2831), 76% of malignant colon carcinomas (Saarnio,
J. et al.,
Am. J. Path., 1997, 153:279-285), 80% of non-small cell lung carcinomas
(Vermylen P. et
al., Eur. Respir. J., 1999, 14:806-811), and in 48% of breast cancers (Chia SK
et al., J.
Clin. Oncol., 2001, 19:3660-3668). Like other tumor associated antigens, the
MN protein
is also present on cells of a limited number of normal tissues, including, for
example,
stomach, bile duct mucosa and the highly proliferate normal cells located in
the small
intestine (Saarnio J. et al., J. Histochem. Cytochem., 1998, 46:497-504).
Human MN cDNA has been cloned and sequenced (Pastorek, et al., Oncogene,
1994, 9:2877-2888). The predicted protein consists of a signal peptide, a
proteoglycan-
related sequence, a carbonic anhydrase domain (carbonic anhydrase IX or CA
IX), a
transmembrane segment, and a short intracellular tail. The carbonic anhydrase
IX domain
catalyzes the reversible hydration of carbon dioxide to carbonic acid. This
activity may
have a role in regulating the local acidification of the extracellular portion
of the tumor
environments, which may consequently lead to the activation of proteases and
finally,
metastasis.
2

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
The regulation of MN expression has also been investigated. In one aspect, for
example, MN expression is up-regulated by hypoxia. The transcriptional complex
known
as hypoxia-inducible factor-i (HIF-1) is a regulator of MN expression.
Accordingly, MN
is known as a HTF-1 responsive gene which has implications in the
understanding of tumor
.5 response to hypoxia (Wykoff CC et al:, Cancer Res., 2000, 60:7075-7083).
Furthermore,
MN expression correlates with tumor hypoxia levels and is a prognostic
indicator of
overall survival and metastasis-free survival in cervical cancer (Loncaster,
JA et al.,
Cancer Res., 2000, 60:7075-7083). IYIN expression also correlates with a high
mean
vessel density, advanced cancer stage, degree of necrosis in head and neck
carcinoma
(Beasley NJP et al., Cancer Res., 2001, 61:5262-5267), poor survival in
nasopharyngeal
carcinoma (Hui EP et al., Clin. Cancer Res., 2002, 8:2595-2604), tumor
necrosis, higher
grade, negative estrogen receptor status, higher relapse rate, and poor
survival for invasive
breast carcinoma (Chia SK et al., J. Clin. Oncol., 2001, 19:3660-3668).
Therefore
expression of MN antigen is correlates with poor survival prognosis, and
cancers of higher
grade.
New and improved therapies for these aggressive cancers, in particular, those
that
target MN expression, are highly desirable and would represent an advancement
in the art.
As such, the present invention discloses new antibody compositions and
immunoconjugates thereof that are useful in the treatment, prevention and/or
diagnosis of
MN-related cancers.
SUMMARY OF THE INVENTION
The present invention relates to antibodies, e.g., monoclonal antibodies, or
antibody fragments that bind to the cell-surface protein MN and which can be
used in the
treatment, prevention and/or diagnosis of cancer. The antibodies of the
invention can
further be conjugated to cytotoxic agents, e.g., monomethylauristatin-E,
and/or co-
3

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
administered or formulated with one or more additional anti-cancer agents. The
anti-MN
antibodies and immunoconjugates of the invention can be used in the methods of
the
invention to treat and/or diagnose and/or monitor cancers, e.g. solid tumors.
In one aspect, the present invention provides an antibody or antibody
fragment, or
a composition that includes the= antibody-or antibody fragment, wherein the
antibody or
fragment has an antigenic binding site that is specifically directed against
an MN protein.
The antigenic binding site may include at least one CDR1, CDR2, or CDR3, or a
CDR1
together with a CDR2 or a CDR3, or a CDR2 together with a CDR 1 or a CDR3, or
a
CDR3 together with a CDRI or CDR 2, or any combination thereof. The CDRI can
be
selected from the group consisting of SEQ ID NOS: 57, 58, 59, 60, 61, 62, 77,
80, 81, 86,
87, 88, 89, 98, 99, 104, 107, and 108. The CDR2 can be selected from the group
consisting of SEQ ID NOS: 63, 64, 65, 66, 67, 68, 69, 78, 82, 83, 90, 91, 92,
93, 100, 101,
105, 109, and 110. The CDR3 can selected from the group consisting of SEQ ID
NOS:
70, 71, 72, 73, 74, 75, 76, 79, 84, 85, 94, 95, 96, 97, 102, 103, 106, 111,
and 112. The
CDR sequences of this aspect of the invention can also include amino acid
sequences that
have preferably greater than about 80% sequence identity, more preferably
greater than
about 85% sequence identity, even more preferably about 90% sequence identity,
still
more preferably about 95% or even about 99% sequence identity, and even up to
about
100% sequence identity to any of the above sequences indicated for each of
CDR1, CDR2
or CDR3.
In another aspect, the antigenic binding site can have a heavy chain variable
region
CDR that is selected from the group consisting of: SEQ ID NOS: 57-85 and an
amino acid
sequence having greater than about 80% sequence identity to any of SEQ ID NOS:
57-85.
4

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
The antigenic binding site can also have a light chain variable region CDR
selected from the group consisting of: SEQ ID NOS: 86-112 and an amino acid
sequence
having greater than about 80% sequence identity to any of SEQ ID NOS: 86-112.
The antigenic binding site can also be selected from a set of specific CDR
sequences that include the following sets of six CDRs:
(a) [3ee9] SEQ ID NOS: 57, 63, 70, 89, 93, and 97;
(b) [3ef2] SEQ ID NOS: 58, 64, 71, 107, 109, and 111;
(c) [1e4] SEQ ID NOS: 59, 65, 72, 107, 109, and 111;
(d) [3a4] SEQ ID NOS: 60, 66, 73, 108, 110, and 112;
(e) [3ab4] SEQ ID NOS: 61, 67, 74, 87, 91, and 95;
(f) [3ah10] SEQ ID NOS: 61, 68, 75, 88, 92, and 96;
(g) [3bb2] SEQ ID NOS: 62, 69, 76, 98, 100, and 102;
(h) [laal] SEQ ID NOS: 77, 78, 79, 86, 90, and 94;
(i) [5a6] SEQ ID NOS: 80, 82, 84, 99, 101, and 103; and
(j) [5aa3] SEQ ID NOS: 81, 83, 85, 104, 105, and 106.
The antigenic binding site may also include a set of heavy chain CDR sequences
selected from the group consisting of:
(a) [3ee9] SEQ IDNOS: 57, 63, and 70;
(b) [3ef2] SEQ ID NOS: 58, 64, and 71;
(c) [le4] SEQ ID NOS: 59, 65, and 72;
(d) [3a4] SEQ ID NOS: 60, 66, and 73;
(e) [3ab4] SEQ ID NOS: 61, 67, and 74;
(f) [3ah10] SEQ ID NOS: 61, 68, and 75;
(g) [3bb2] SEQ ID NOS: 62, 69, and 76;
(h) [laal] SEQ ID NOS: 77, 78, and 79;
5

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
(i) [5a6] SEQ IDNOS: 80, 82, and 84; and
(j) [5aa3] SEQ ID NOS: 81, 83, and 85.
The antigenic binding site can also include a set of light chain CDR sequences
selected from the group consisting of:
(a) [3ee9] SEQ IDNOS: 89, 93; and 97; = -
(b) [3ef2] SEQ ID NOS: 107, 109, and 111;
(c) [1e4] SEQ ID NOS: 107, 109, and 111;
(d) [3a4] SEQ ID NOS: 108, 110, and 112;
(e) [3ab4] SEQ ID NOS: 87, 91, and 95;
(f) [3ah10] SEQ ID NOS: 88, 92, and 96;
(g) [3bb2] SEQ ID NOS: 98, 100, and 102;
(h) [laal] SEQ ID NOS: 86, 90, and 94;
(i) [5a6] SEQ ID NOS: 99, 101, and 103; and
(j) [5aa3] SEQ ID NOS: 104, 105, and 106.
In yet another aspect, the present invention provides an antibody or antibody
fragment that has an antigenic binding site that contains a pair of heavy
chain variable and
light chain variable regions selected from the group consisting of:
(a) the heavy chain variable region of SEQ ID NO: 133 and the light chain
variable region of SEQ ID NO: 134;
(b) the heavy chain variable region of SEQ ID NO:135 and the light chain
variable region of SEQ ID NO:136;
(c) the heavy chain variable region of SEQ ID NO: 137 and the light chain
variable region of SEQ ID NO:138;
(d) the heavy chain variable region of SEQ ID NO:139 and the light chain
variable region of SEQ ID NO: 140;
6

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
(e) the heavy chain variable region of SEQ ID NO:141 and the light chain
variable region of SEQ ID NO: 142;
(f) the heavy chain variable region of SEQ ID NO: 143 and the light chain
variable region of SEQ ID NO: 144;
(g) the heavy chain variable region of SEQ ID NO: 145 and the light chain
variable region of SEQ ID NO: 146;
(h) the heavy chain variable region of SEQ ID NO: 147 and the light chain
variable region of SEQ ID NO:148;
(i) the heavy chain variable region of SEQ ID NO: 149 and the light chain
variable region of SEQ ID NO: 150; and
(j) the heavy chain variable region of SEQ ID NO:151 and the light chain
variable region of SEQ ID NO:152.
The antibodies or antibody fragments of the invention can bind to the MN
protein
with a dissociation constant of preferably about 0.15 nM to about 50 nM.
In another aspect, the antibodies or fragments of the invention are IgG
antibodies
or IgG fragments. The antibodies or fragments can also be IgG1, IgG2a, IgG2b,
IgG3,
IgM, IgD, IgE, IgA, or IgM antibodies, Fab fragments, F(ab')2 fragments, scFv
fragments,
Fv fragments, a diabodies, linear antibodies, single-chain antibodies,
biospecific
antibodies, multispecific antibodies, or chimeric antibodies (e.g. comprising
a human
antibody scaffold grafted to a human or non-human antibody binding region, or
a non-
human antibody scaffold grafted to a human or non-human antibody binding
region). The
chimeric antibodies can include, for example, antibody scaffold regions from
non-human
sources, such as, for example, cow, mouse, ilanza, camel, or rabbit. Further
information
on the engineering of antibodies can be found in the literature, for example,
Holliger and
Hudson, Nature Biotechnology, (Sep, 2005) 23:1126-1136, which is incorporated
herein
7

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
by reference. The aforementioned fragments can be obtained from an
immunoglobulin or
produced by a suitable means, e.g. recombinant expression, in a fragment form.
The antibodies or antibody fragments of the invention can also be humanized,
wherein the CDR sequences or regions (e.g. CDR 1, CDR2, CDR3) can be non-
human,
e.g. murine.
The antibodies or antibody fragments of the invention, or compositions
including
the antibodies or fragments, can include a cytoxic agent that is conjugated to
the antibody
or fragment. In one aspect, the cytotoxic agent is monomethylauristatin-E
(MMAE),
however, other cytoxic agents are also provided, which can include, for
example,
functional analogs of MMAE (e.g. monomethylauristatin-F), and other cytotoxic
agents,
e.g., aplidin, azaribine, anastrozole, azacytidine, bleomycin, bortezomib,
bryostatin-1,
busulfan, calicheamycin, camptothecin, 10-hydroxycamptothecin, carmustine,
celebrex,
chlorambucil, cisplatin, irinotecan (CPT-1 1), SN-38, carboplatin, cladribine,
cyclophosphamide, cytarabine, dacarbazine, docetaxel, dactinomycin, daunomycin
glucuronide, daunorubicin, dexamethasone, diethylstilbestrol, doxorubicin,
doxorubicin
glucuronide, epirubicin glucuronide, ethinyl estradiol, estramustine,
etoposide, etoposide
glucuronide, etoposide phosphate, floxuridine (FUdR), 3',5'-O-dioleoyl-FudR
(FUdR-dO),
fludarabine, flutamide, fluorouracil, fluoxymesterone, gemcitabine,
hydroxyprogesterone
caproate, hydroxyurea, idarubicin, ifosfamide, L-asparaginase, leucovorin,
lomustine,
mechlorethamine, medroprogesterone acetate, megestrol acetate, melphalan,
mercaptopurine, 6-mercaptopurine, methotrexate, mitoxantrone, mithramycin,
mitomycin,
mitotane, phenyl butyrate, prednisone, procarbazine, paclitaxel, pentostatin,
PSI-341,
semustine streptozocin, tamoxifen, taxanes, taxol, testosterone propionate,
thalidomide,
thioguanine, thiotepa, teniposide, topotecan, uracil mustard, velcade,
vinblastine,
vinorelbine, vincristine, ricin, abrin, ribonuclease, onconase, rapLRl, DNase
I,
8

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria
toxin,
Pseudomonas exotoxin, and Pseudoinonas endotoxin, or combinations thereof. Any
of the
cytoxic agents can also include functional analogs thereof.
The compositions of the invention can include in addition to the antibodies
and
fragments (with or without the aforementioned conjugated cytoxic agents)
various anti-
cancer agents, which can include, for example, bleomycin, docetaxel
(Taxotere),
doxorubicin, edatrexate, erlotinib (Tarceva), etoposide, finasteride
(Proscar), flutamide
(Eulexin), gemcitabine (Gemzar), genitinib (Irresa), goserelin acetate
(Zoladex),
granisetron (Kytril), imatinib (Gleevec), irinotecan (Campto/Camptosar),
ondansetron
(Zofran), paclitaxel (Taxol), pegaspargase (Oncaspar), pilocarpine
hydrochloride
(Salagen), porfimer sodium (Photofrin), interleukin-2 (Proleukin), rituximab
(Rituxan),
topotecan (Hycamtin), trastuzumab (Herceptin), Triapine, vincristine, and
vinorelbine
tartrate (Navelbine), or therapeutic antibodies or fragments thereof, or anti-
angiogenic
agent, such as, for example, angiostatin, bevacizumab (Avastin ), sorafenib
(Nexavar ),
baculostatin, canstatin, maspin, anti-VEGF antibodies or peptides, anti-
placental growth
factor antibodies or peptides, anti-Flk-1 antibodies, anti-Flt-1 antibodies or
peptides,
laminin peptides, fibronectin peptides, plasminogen activator inhibitors,
tissue
metalloproteinase inhibitors, interferons, interleukin 12, IP-l0, Gro-0,
thrombospondin, 2-
methoxyoestradiol, proliferin-related protein, carboxiamidotriazole, CM101,
Marimastat,
pentosan polysulphate, angiopoietin 2, interferon-alpha, herbimycin A,
PNU145156E,
16K prolactin fragment, Linomide, thalidomide, pentoxifylline, genistein, TNP-
470,
endostatin, paclitaxel, accutin, cidofovir, vincristine, bleomycin, AGM-1470,
platelet
factor 4 or minocycline.
The present invention further provides in another aspect a method for treating
an
MN-related disorder by administering a therapeutically effective amount of the
antibodies
9

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
and/or fragments of the invention, or the compositions of the invention which
include the
antibodies and/or fragments of the invention. The MN-related disorder can
include, for
example, cancer, such as, a solid tumor cancer. The solid tumor can be in or
originating
from the breast, respiratory tract, lung, brain, reproductive organ, digestive
tract, colon,
urinary tract, kidney, esophagus, cervix, eye, liver, skin, head, neck,
thyroid, and
parathyroid.
In another aspect, the present invention provides a method of diagnosing an MN-
related disorder characterized abnormal MN levels comprising comparing the
level of MN
in a suspected diseased tissue or cell with the level of MN in a corresponding
healthy
tissue or cell, wherein an abnormal MN level in the suspected diseased tissue
or cell is an
indication of an MN-related disorder, said step of comparing further
comprising detecting
by immunoassay the level of MN in the diseased tissue and the healthy tissue
with the
antibodies or antibody fragments of the invention.
In a particular aspect, the invention provides a method of diagnosing an MN-
related disorder where the immunoassay includes the steps of: (a) detecting
the level of
MN protein in the healthy tissue; (b) detecting the level of MN protein in the
suspected
diseased tissue; and (c) comparing the levels of MN protein from (a) and (b).
An elevated
level of MN protein in the suspected diseased tissue as compared to the level
of MN
protein in the healthy tissue is indicative of the presence of an MN-related
disorder.
Also provided by the present invention is a kit comprising the antibodies or
antibody fragments of the invention, or alternatively, the compositions of the
invention,
and a set of instructions for using the kit in a method of treating an MN-
related disorder or
for diagnosing a an MN-related disorder.
These and other embodiments are disclosed or are obvious from and encompassed
by, the following Detailed Description.

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
BRIEF DESCRIPTION OF THE DRAWINGS
The following detailed description, given by way of example, but not intended
to
limit the invention solely to the specific embodiments described, may best be
understood
in conjunction with the accompanying drawings, in which:
Figure I depicts DNA sequences of the antibody complementarity determining
regions (CDRs);
Figure 2 depicts the protein sequences of the antibody complementarity
determining regions (CDRs);
Figure 3 depicts the DNA sequences of specific antibody light and heavy chain
variable regions, each of which contains both CDRs and framework regions;
Figure 4 depicts protein sequences of specific antibody light (VL) and heavy
(VH)
chain variable regions, each of which contains both the CDRs and the framework
regions;
Figure 5 depicts the MN -binding properties for the MN antibodies of the
present
invention;
Figure 6 depicts the prevention of tumor cell adhesion to MN coated plates
produced by incubation with an anti-MN antibody;
Figure 7 depicts the in vivo anti-tumor activity in a xenograft model
comprising the
MaTu tumor resulting from treatment with an immunoconjugate comprising the
anti-MN
monoclonal antibody 1e4;
Figure 8 depicts the FACS measurement of binding of an anti-MN antibody to the
PC3mm2 cell line which expresses the MN protein on its surface;
Figure 9 schematically depicts the antibody conjugate (anti-MN antibody
conjugated to MMAE);
Figure l0a shows FACS plots depicting 3ee9/MMAE binding to MN+ (MaTu)
cells, but not to MN- (DLD) cells;
11

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Figure lOb shows FACS plots depicting lE4 immunoconjugate binding to MN+
(PC3mm2) cells, but not to MN- (DLD) cells;
Figure lOc shows FACS plots depicting laal immunoconjugate binding to MN+
(PC3mm2) cells, but not to MN- (DLD) cells;
Figure 1 la shows immunofluorescence images depicting internalization of
3ee9/MMAE by MN+ cells and lack of internalization by MN- cells. Internalized
3ee9/MMAE is shown as fluorescence;
Figure 1lb shows immunofluorescence images depicting internalization of the
1.E4
immunoconjugate by MN+ cells and lack of internalization by MN- cells.
Internalized
lE4 inununoconjugate is shown as fluorescence;
Figure 12 shows an immunoblot depicting specific immunoprecipitation of
biotinylated cell surface proteins by MN antibodies;
Figure 13a graphically depicts the cytotoxicity of 3ee9IMMAE against MN+, but
against MN-, cells;
Figure 13b graphically depicts the cytotoxicity of the 1E4 immunoconjugate
against MN+, but against MN-, cells;
Figure 13c graphically depicts the cytotoxicity of the laal immunoconjugate
against MN+, but against MN-, cells;
Figure 14 shows immunofluorescence images depicting 3ee9/MMAE's prevention
of normal spindle formation by tubulin inhibition;
Figure 15 graphically depicts the anti-tumor efficacy of 3ee9/MMAE against
MaTu xenografts;
Figure 16 graphically depicts the anti-tumor efficacy of the 1E4
immunoconjugate
against established MaTu breast tumors;
12

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Figure 17 graphically depicts the anti-tumor efficacy of the laal
immunoconjugate
against MaTu xenografts;
Figure 18 graphically depicts the therapeutic index (TI) of 3ee9/MMAE against
MaTu xenografts;
Figure 19 graphically depicts the anti-tumor efficacy of 3ee9/MMAE and free
MMAE against HT-29 xenografts;
Figure 20 graphically depicts the anti-tumor efficacy of 3ee9/MMAE against HT-
29 xenografts, following the Q7Dx2 schedule;
Figure 21 graphically depicts the anti-tumor efficacy of 3ee9/MMAE against HT-
29 xenografts, following the Q lDx 1 schedule;
Figure 22 graphically depicts the anti-tumor efficacy of 3ee9/MMAE against
PC3mm2 xenografts;
Figure 23 graphically depicts the anti-tumor efficacy of 3ee9/MMAE against
Colo-
205 xenografts;
Figure 24 graphically depicts the anti-tumor efficacy of 3ee9/MMAE against HCT-
15 xenografts;
Figure 25 graphically depicts the anti-tumor efficacy of 3ee9/MMAE against MN-
(in left-hand graph) and MN+ (in right-hand graph) MIAPaca2 xenografts;
Figures 26a and 26b graphically depict the anti-tumor efficacy of 3ee9/MMAE in
combination with Xeloda against Colo-205 CRC xenografts at varying doses;
Figure 27 shows immunofluorescence images depicting in vivo localization of
3ee9
in huMN- MIAPaca2 and MIAPaca2 tumors; and
Figure 28 show immunofluorescence images (of histological samples) depicting
3ee9/MMAE inhibition of tubulin polymerization in HT-29 CRC xenografts at two
doses.
13

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Figure 29 shows the complete nucleotide sequence of the insert region of the
mammalian expression vector 3ee9H+,.pCMVucoF8 (see Example 21) which encodes a
human IgG anti-MN antibody comprising the kappa and heavy CDR variable regions
of
SEQ ID NOS: 126 and 125, respectively, obtained from vector 3ee9pMORPHx9 (see
Examples 1-3). SEQ ID NO: 153.
DETAILED DESCRIPTION OF THE INVENTION
It is to be understood that present invention as described herein is not to be
limited
to the particular details set forth herein regarding any aspect of the present
invention,
including, anti-MN antibodies, immunoconjugates, methods of treatment,
protocols, cell
lines, animal species or genera, constructs, and reagents described and, as
such, may vary.
It is also to be understood that the terminology used herein is for the
purpose of describing
particular embodiments only, and is not intended to limit the scope of the
present
invention.
Definitions
Unless defined otherwise, all technical and scientific terms used herein have
the
meaning commonly understood by one of ordinary skill in the art to which this
invention
belongs. The following references, however, can provide one of skill in the
art to which
this invention pertains with a general definition of many of the terms used in
this
invention, and can be referenced and used so long as such definitions are
consistent the
meaning commonly understood in the art. Such references include, but are not
limited to,
Singleton et al., Dictionary of Microbiology and Molecular Biology (2d ed.
1994); The
Cambridge Dictionary of Science and Technology (Walker ed., 1988); Hale &
Marham,
The Harper Collins Dictionary of Biology (1991); and Lackie et al., The
Dictionary of
Cell & Molecular Biology (3d ed. 1999); and Cellular and Molecular Immunology,
Eds.
Abbas, Lichtman and Pober, 2 d Edition, W.B. Saunders Company. Any additional
14

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
technical resources available to the person of ordinary skill in the art
providing definitions
of terms used herein having the meaning commonly understood in the art can be
consulted. For the purposes of the present invention, the following terms are
further
defined. Additional terms are defined elsewhere in the description.
As used herein and=in the appended claims, the singular forms "a," "and," and
"the" include plural reference unless the context clearly dictates otherwise.
Thus, for
example, reference to "a gene" is a reference to one or more genes and
includes
equivalents thereof known to those skilled in the art, and so forth.
As used herein, the term "antibody" includes immunoglobulin molecules (e.g.,
any
lo type, including IgG, IgE, IgM, IgD, IgA and IgY, and/or any class,
including, IgGl, IgG2,
IgG3, IgG4, IgA 1 and IgA2) isolated from nature or prepared by recombinant
means.
Antibodies also are meant to encompass antigen-binding antibody fragments,
such as Fab,
F(ab')2, scFv (single chain Fvs), Fv, single chain antibodies, diabodies,
disulfide-linked
Fvs (sdFv), and fragments comprising a VL or VH domain, which are prepared
from intact
immunoglobulins or prepared by recombinant means.
The antibodies and/or antigen-binding antibody fragments of the present
invention
may be monospecific (e.g. monoclonal), bispecific, trispecific or of greater
multi
specificity. Multispecific antibodies may be specific for different epitopes
of an antigen or
may be specific for epitopes of more than one antigen. See, e.g., PCT
publications WO
93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., 1991, J.
Immunol.
147:60 69; U.S. Pat. Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920;
5,601,819;
Kostelny et al., 1992, J. Immunol. 148:1547 1553, each of which are
incorporated herein
by reference.
Antigen-binding antibody fragments may comprise the variable region(s) alone
or
in combination with the entirety or a portion of the following: hinge region,
CH1, CH2,

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
CH3 and CL domains. Also included in the invention are antigen-binding
antibody
fragments also comprising any combination of variable region(s) with a hinge
region,
CH1, CH2, CH3 and CL domain.
Preferably, the antibodies or antigen-binding antibody fragments are human,
humanized, murine ~e.g., mouse and rat); donkey, sheep, rabbit, goat, guinea
pig, camelid,
horse, or chicken. As used herein, "human" antibodies include antibodies
having the
amino acid sequence of a human immunoglobulin and include antibodies isolated
from
human imrnunoglobulin libraries, from human B cells, or from animals
transgenic for one
or more human immunoglobulin, as described infra and, for example in U.S. Pat.
No.
5,939,598 by Kucherlapati et al. The term antibody also extends to other
protein scaffolds
that are able to orient antibody CDR inserts into the same active binding
conformation as
that found in natural antibodies such that binding of the target antigen
observed with these
chimeric proteins is maintained relative to the binding activity of the
natural antibody from
which the CDRs were derived.
1s As used herein, the term "humanized" forms of non-human (e.g., murine)
antibodies are chimeric antibodies which contain minimal sequence derived from
non-
human immunoglobulin. For the most part, humanized antibodies are human
immunoglobulins (recipient antibody) in which hypervariable region residues
(e.g. the
complementarity determining regions "CDR") of the recipient are replaced by
hypervariable region residues (CDRs) from a non-human species (donor antibody)
such as
mouse, rat, rabbit, or nonhuman primate having the desired specificity,
affinity, and
capacity. In some instances, framework region (FR) residues of the human
immunoglobulin may be replaced by corresponding non-human residues.
Furthermore,
humanized antibodies may comprise residues which are not found in the
recipient
antibody or in the donor antibody. Such modifications are made to further
refine antibody
16

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
performance. In general, the humanized antibody may comprise substantially all
of at
least one or typically two variable domains, in which all or substantially all
of the
hypervariable regions correspond to those of a non-human immunoglobulin and
all or
substantially all of the FRs are those of a human immunoglobulin sequence. The
-5 humanized antibody optionally also may comprise-at least a portion of an
immunoglobulin
constant region (Fc), typically that of a human immunoglobulin. For a review,
see Jones,
et al., (Nature 321:522-525, 1986); Reichmann, et al., (Nature 332:323-329,
1988); and
Presta, (Curr. Op. Struct. Biol. 2:593-596, 1992). The preparation of
humanized
antibodies can be found in U.S. Patent Nos. 7,049,135, 6,828,422, 6,753,136,
6,706,484,
6,696,248, 6,692,935, 6,667,150, 6,653,068, 6,300,064, 6,294,353, and
5,514,548, each of
which are incorporated herein in their entireties_
As used herein, the term "single-chain Fv" or "sFv" antibody fragments
comprise
the VH and VL domains of an antibody, wherein these domains are present in a
single
polypeptide chain. Generally, the Fv polypeptide further comprises a
polypeptide linker
between the VH and VL domains which enables the sFv to form the desired
structure for
antigen binding. For a review, see Pluckthun (The Pharmacology of Monoclonal
Antibodies, Vol. 113, Rosenburg and Moore eds. Springer-Verlag, New York, pp.
269-
315, 1994), which is incorporated herein in its entirety by reference.
The term "diabodies" refers to small antibody fragments with two antigen-
binding
sites, which fragments comprise a heavy chain variable domain (VH) connected
to a light
chain variable domain (VL) in the same polypeptide chain (VH-VL). By using a
linker that
is too short to allow pairing between the two domains on the same chain, the
domains are
forced to pair with the complementary domains of another chain and create two
antigen-
binding sites. Diabodies are described more fully in, for example, EP 404,097;
WO
17

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
93/11161; and Hollinger, et al., (Proc. Natl. Acad. Sci. USA 90:6444-6448,
1993), each of
which are incorporated by reference.
The expression "linear antibodies" refers to the antibodies described in the
art, for
example, in Zapata, et al., (Protein Eng. 8(10):1057-1062, 1995), which is
incorporated by
reference. Briefly, such-antibodies. comprise a pair of tandem Fd segments (VH-
CHI-VH-
Cy1) which form a pair of antigen binding regions. Linear antibodies can be
bispecific or
monospecific.
The term "monoclonal antibody" as used herein refers to an antibody obtained
from a population of substantialIy homogeneous antibodies, that is, individual
antibodies
comprising an identical population except for possible naturally occurring
mutations that
may be present in minor amounts. Monoclonal antibodies are highly specific,
that is,
directed against a single antigenic site. Furthermore, in contrast to
conventional
(polyclonal) antibody preparations which typically include different
antibodies directed
against different determinants (epitopes), each monoclonal antibody is
directed against a
single determinant on the antigen. The modifier "monoclonal" indicates the
character of
the antibody as being obtained from a substantially homogeneous population of
antibodies, and is not to be construed as requiring production of the antibody
by any
particular method. For example, the monoclonal antibodies to be used in
accordance with
the present invention may be made by the hybridoma method first described by
Kohler, et
al., (Nature 256:495, 1975), or may be made by recombinant DNA methods (see,
e.g.,
U.S. Patent No. 4,816,567). Monoclonal antibodies may also be isolated from
phage
antibody libraries using the techniques described in, for example, Clackson,
et al., (Nature
352:624-628,1991) and Marks, et al., (J. Mol. Biol. 222:581-597, 1991).
The monoclonal antibodies herein also include "chimeric" antibodies in which a
portion of the heavy and/or light chain is identical with or homologous to
corresponding
18

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
sequences in antibodies derived from a particular species or belonging to a
particular
antibody class or subclass, while the remainder of the chain(s) is identical
with or
homologous to corresponding sequences in antibodies derived from another
species or
belonging to another antibody class or subclass, as well as fragments of such
antibodies,
so long as they exhibit the desired biological= activity (see, e.g., U.S.
Patent No. 4,816,567;
and Morrison, et al., Proc. Natl. Acad. Sci. USA 81:6851-6855, 1984, each of
which are
incorporated by reference).
As used herein, the terms "biological sample" or "patient sample" as used
herein,
refers to a sample obtained from an organism or from components (e.g., cells)
of an
organism. The sample may be of any biological tissue or fluid. The sample may
be a
"clinical sample" which is a sample derived from a patient. Such samples
include, but are
not limited to, sputum, blood, serum, plasma, blood cells (e.g., white cells),
tissue samples,
biopsy samples, urine, peritoneal fluid, and pleural fluid, saliva, semen,
breast exudate,
cerebrospinal fluid, tears, mucous, lymph, cytosols, ascites, amniotic fluid,
bladder
washes, and bronchioalveolar lavages or cells therefrom, among other body
fluid samples.
The patient samples may be fresh or frozen, and may be treated with heparin,
citrate, or
EDTA. Biological samples may also include sections of tissues such as frozen
sections
taken for histological purposes.
The term "cancer" includes, but is not limited to, solid tumors, such as
cancers of
the breast, respiratory tract, brain, reproductive organs, digestive tract,
urinary tract, eye,
liver, skin, head and neck, thyroid, parathyroid, and their distant
metastases. The term
also includes sarcomas, lymphomas, leukemias, and plasma cell myelomas.
Examples of breast cancer include, but are not limited to, invasive ductal
carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular
carcinoma in
situ. Examples of cancers of the respiratory tract include, but are not
limited to, small-cell
19

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
and non-small-cell lung carcinoma, as well as bronchial adenoma and
pleuropulmonary
blastoma. Examples of brain cancers include, but are not limited to, brain
stem and
hypophtalrnic glioma, cerebellar and cerebral astrocytoma, medulloblastoma,
ependymoma, as well as neuroectodermal and pineal tumor. Tumors of the male
reproductive organs include, but are =not limited to, prostate and testicular-
cancer. Tumors
of the female reproductive organs include, but are not lirnited to,
endometrial, cervical,
ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus. Tumors
of the
digestive tract include, but are not limited to, anal, colon, colorectal,
esophageal,
gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland
cancers. Tumors
of the urinary tract include, but are not limited to, bladder, penile, kidney,
renal pelvis,
ureter, and urethral cancers. Eye cancers include, but are not limited to,
intraocular
melanoma and retinoblastoma. Examples of liver cancers include, but are not
limited to,
hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar
variant),
cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed
hepatocellular
cholangiocarcinoma. Skin cancers include, but are not limited to, squamous
cell
carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and
non-
melanoma skin cancer. Head-and-neck cancers include, but are not limited to,
laryngeal /
hypopharyngeal / nasopharyngeal / oropharyngeal cancer, and lip and oral
cavity cancer.
Lymphomas include, but are not limited to, AIDS-related lymphoma, non-
Hodgkin's
lymphoma, cutaneous T-cell lymphoma, Hodgkin's disease, and lymphoma of the
central
nervous system. Sarcomas include, but are not limited to, sarcoma of the soft
tissue,
osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and
rhabdomyosarcoma.
Leukemias include, but are not limited to, acute myeloid leukemia, acute
lymphoblastic
leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and
hairy cell
leukemia.

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
As used in this invention, the term "epitope" means any antigenic determinant
on
an antigen, e.g. MN protein, to which the antibody binds through an antigenic
binding site.
Determinants or antigenic determinants usually consist of chemically active
surface
groupings of molecules such as amino acids or sugar side chains and usually
have specific
three dimensional structural characteristics; as well as specific charge
characteristics.
The term "specifically immunoreactive" refers to a binding reaction between an
antibody and a protein, compound, or antigen, having an epitope recognized by
the
antigenic binding site of the antibody. This binding reaction is determinative
of the
presence of a protein, antigen or epitope having the recognized epitope
amongst the
presence of a heterogeneous population of proteins and other biologics. In the
context of
an imrnunoassay, specifically immunoreactive antibodies can bind to a protein
having the
recognized epitope and bind, if at all, to a detectably lesser degree to other
proteins lacking
the epitope which are present in the sample. In an in vivo context,
"specifically
immunoreactive" can refer to the conditions under which in an animal forms an
immune
response against a vaccine or antigen, e.g. a humoral response to the antigen
(the
production of antibodies, against a vaccine, protein, compound, or antigen
presented
thereto under immunologically reactive conditions) or a cell-mediated (also
herein as
"cellular immune response", i.e. a response mediated by T lymphocytes against
the
vaccine, protein, compound or antigen presented thereto).
As used herein, the term "immunologically reactive conditions" is used in the
context of an immunoassay or an in vitro reaction wherein the physical
conditions of the
reaction, including, for example, the temperature, salt concentration, pH,
reagents and
their concentrations, and the concentrations of antigen and cognate antibody
that is
specifically immunoreactive to the antigen, are provided or adjusted to allow
binding of
the cognate antibody to the antigen. Immunologically reactive conditions are
dependent
21

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
upon the format of the antibody binding reaction and, typically are those
utilized in
immunoassay protocols. See Harlow and Lane (1988) Antibodies: A Laboratory
Manual,
Cold Spring Harbor Publications, New York, for a description of immunoassay
formats
and conditions.
.=5 The term "patient" or "subject" as used herein includes mammals (e.g.,
humans
and animals).
Antibodies
The present invention relates to antibodies that bind to MN. These antibodies
may
be useful for a variety of therapeutic and diagnostic purposes.
It will be generally appreciated by those skilled in the art that the most
critical
determinants of antibody selectivity and binding affinity are the sequences
and resulting
conformations of the complementarity regions (CDRs). Most antibodies contain
six
CDRs, three within the heavy chain variable region (VH) and three within the
light chain
variable region (VL). The intervening sequences between the CDRs within VH and
VL
are the framework regions which orient the CDRs. The CDRs together form the
antigenic
binding sites within antibodies. The critical role of these CDRs in
determining the
functional properties of antibodies has long been exploited in the processes
of antibody
humanization and antibody optimization_ In the former process, the CDRs from a
monoclonal antibody, for example, a mouse antibody are transferred to a human
antibody
of similar framework design thereby resulting in an antibody with the same
functional
properties and reduced immunogencity in man.
The success of this process is evident from the number of humanized antibodies
that have been successfully commercialized as human therapeutics and include
Herceptin (trastuzumab, Genentech, Inc., South San Francisco, CA), Synagis0
(palivizumab, Medimmune, Inc., Gaithersburg, MD), Campath0 (alemtuzumab,
Genzyme
22

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Oncology, Cambridge, MA), Zenapax0 (daclizumab, Roche Pharmaceuticlas, Nutley,
NJ), Xolair0 (omalizumab, Genentech, Inc., South San Francisco, CA), Raptiva
(efalizumab, Genentech, Inc., South San Francisco, CA), Avastin (bevacizumab,
Genentech, Inc., South San Francisco, CA), and Mylotarg0 (gemtuzumab
ozogamicin,
Wyeth-Ayerst, Madisoni- NJ): Other examples have been described in Singer, et
al., (J.
Inununol, 150:2844-2857, 1993); Luo, et al., (J. Immunol Meth., 275:31-40,
2002); and
Kostelny, et al., (Int. J. Cancer 93;556-565, 2001).
The process of antibody optimization focuses on improvements in antibody
selectivity or binding affinity through specific alteration in the sequence of
the CDRs. It is
well accepted within the field of antibody development that the CDRs encode
binding
affinity and selectivity properties required for therapeutic and diagnostic
uses, and these
CDR sequences may be used to confer such desirable functional properties to a
wide
variety of alternate antibody frameworks using standard procedures known to
those skilled
in the art. It is also possible to transfer antibody binding activity by
grafting antibody
CDRs onto other proteins that possess immunoglobulin-like folds such as other
proteins
within the immunoglobulin superfamily and non-immunoglobulins with sirnilar
folds to
immunoglobulins (Nicaise, et al., Protein Science 13:1882-1891, 2004).
The present invention contemplates any known or suitable technology for the
preparation of the antibodies and antigen binding antibody fragments of the
invention.
For example, phage display technology is useful for obtaining high affinity
anti-
MN antibodies or antigen binding antibody fragments of the invention for the
use in
diagnosis and/or treatment of an MN-related disorder, such as, an MN-related
cancer. The
technology, referred to as affinity maturation, employs mutagenesis or CDR
walking and
re-selection using the MN antigen to identify antibodies that bind with higher
affinity to
the antigen when compared with the initial or parental antibody (see, e.g.,
Glaser et al.,
23

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
1992, J. Immunology 149:3903). Mutagenizing entire codons rather than single
nucleotides results in a semi-randomized repertoire of amino acid mutations.
Libraries can
be constructed consisting of a pool of variant clones each of which differs by
a single
amino acid alteration in a single CDR and which contain variants representing
each
possible amino acid substitution for each CDR residue. Mutants with increased
binding
affinity for the antigen can be screened by contact with the immobilized
mutants
containing labeled antigen. Any screening method known in the art can be used
to
identify mutant antibodies with increased avidity to the antigen (e.g., ELISA)
(See Wu et
al., 1998, Proc Natl. Acad. Sci. USA 95:6037; Yelton et al., 1995, J.
Inununology
155:1994, incorporated by reference). CDR walking may also be used to
randomize the
light chain (See Schier et al., 1996, J. Mol. Bio. 263:551, incorporated by
reference).
In a particular example, MorphoSys AG (Germany) provides a phage-antibody
technology that may be used to generate fully human antibodies. The Morphosys
HuCAL
GOLD library provides a number of advancements over earlier versions of the
technology
(Knappik, et al., J. Mol. Biol. 296:57-86, 2000, incorporated by reference)
including the
HuCAL-Fab 1 library described in Rauchenberger, et al., (J. Biol. Chem.
278:38194-
38205, 2003, incorporated by reference)_ Like earlier versions, HuCAL GOLD
incorporates a human antibody design, that features human consensus framework
sequences and patterns of CDR variability that mimic the natural human
sequence
diversity. However, diversity is extended in HuCAL GOLD to include all six
antibody
CDRs. Moreover, recovery of high affinity antibodies is augmented through the
CysDisplayTM feature (Kretzschmar and von Ruden, Curr. Opin. Biotechnol.
13:598-602,
2002). Antibodies derived with this technology exhibit a greatly reduced
probability of
immunogenicity.
24

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
The phage-display technologies, such as those available from Morphosys, are
known in the art and reference can further be made to Boehncke WH et al., Br J
Derrnatol.
(2005), Nov;153(5): 1092; Simon Moroney et al., Modern Biopharmaceuticals;
Edited by
J. Knaeblein, Wiley-VCH Verlag (2005); Ralf Ostendorp et al., Antibodies,
Volume 2:
== 5 Novel technologies- and. therapeutic use, p.13-52, (2004), Kluwer
Acadenuic/Plenum
Publishers, New York; R. Rauchenberger et al., J Biol Chem., (2003), Oct 3,
278(40):
38194-38205; T. Kretzschmar et al., Cun Opin Biotechnol., (2002), Dec,
13(6):598-602;
Krebs B et al, J Irnmunol Methods, (2001), Aug 1, 254(1-2); M. Marget et al.,
Tissue
Antigens, (2000), 56: 1-9; A. Knappik et al., J. Mol Biol., (2000), Feb 11,
296 (1): 57-86;
and A. Pluckthun et al., Immunotechnology, (1997), Jun; 3(2): 83-105, each of
which are
incorporated herein by reference in their entireties.
As an example, antibodies with MN binding and cell adhesion-neutralizing
activity
may be identified using the MorphoSys technology. The MN protein may be coated
on
microtiter plate or a magnetic bead and incubated with the MorphoSys HuCAL-
GOLD
Fab phage library. Phage-linked Fabs that do not bind to MN may be washed from
the
plate, leaving only phage that bind to MN. The bound phage may be eluted by
addition of
a thiol reducing agent such as dithiotreitol (DTT) resulting in cleavage of
the disulfide
bond linking the antibody to the phage. The recovered population of phage may
be
enriched with phage expressing MN binding antibody fragments and may be
amplified by
infection of E. coli hosts. This panning process may be repeated using the
enriched
population of phage to further enrich for a population of phage-linked
antibodies that bind
to MN. The gene sequence encoding the Fabs may then be excised using standard
cloning
techniques and transferred to an expression vector, such as a bacterial (e.g.,
E. coli)
expression vector, or a mammalian expression vector, which is used to
transform an host

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
cell line, such as, a CHO or E. coli expression cell line. Fabs from the
enriched pool may
then be expressed and purified.
Alternatively, panning may be performed using MN expressing cells as antigen.
For example, cells transfected with MN antigen may be labeled with biotin.
These
= 5 transfected cells may then be -mixed with unlabeled, non-MN transfected
cells at a labeled
to unlabeled ratio of 1:10. The phage library is added to the cells, and the
biotinylated,
MN-bearing cells are captured with streptavidin-bound magnetic beads that are
bound to a
magnet. Non-specific phage are washed away, and the MN-bearing cells are
specifically
eluted by removing the magnetic field. These specifically bound phage may be
amplified
for further rounds of cell panning or may be alternated with peptide and/or
protein
panning.
Antibodies may be produced by a variety of other techniques as described
below.
For example, another approach for obtaining antibodies is to screen a DNA
library from B
cells as described by Dower, et al., (WO 91/1727 1, incorporated by reference)
and
McCafferty, et al., (WO 92/01047) (each of which is incorporated by reference
in its
entirety). In these methods, libraries of phage are produced in which members
display
different antibodies on their outer surfaces. Antibodies are usually displayed
as Fv or Fab
fragments. Phage displaying antibodies are selected by affinity enrichment for
binding to
a selected protein.
In a variation of the phage-display method, human antibodies having the
binding
specificity of a selected murine antibody may be produced (e.g., WO 92/20791,
incorporated by reference). In this method, either the heavy or light chain
variable region
of the selected murine antibody (e.g., 5C7.29) may be used as a starting
material. If, for
example, a light chain variable region is selected as the starting material, a
phage library
may be constructed in which members displays the same light chain variable
region (i.e.,
26

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
the murine starting material) and a different heavy chain variable region. The
heavy chain
variable regions may be obtained from a library of rearranged human heavy
chain variable
regions. A phage showing strong specific binding for a protein (e.g., at least
10 nM or at
least 1 nM) is selected. The human heavy chain variable region from this phage
then
=5 serves as a starting material for constructing a further phage library. In
this library, each
phage displays the same heavy chain variable region (i.e., the region
identified from the
first display library) and a different light chain variable region. The light
chain variable
regions may be obtained from a library of rearranged human variable light
chain regions.
Again, phage showing strong specific binding are selected.
As another example, antibodies may also be produced using trioma methodology.
The basic approach and an exemplary cell fusion partner, SPAZ-4 for use in
this approach,
have been described by Oestberg, et al., (Hybridoma 2:361-367, 1983; U.S.
Patent No.
4,634,664, each incorporated by reference); and Engleman, et al., (U.S. Patent
No.
4,634,666, incorporated by reference). The antibody-producing cell lines
obtained by this
method are called triomas, because they are descended from three cells--two
human and
one mouse. Initially, a mouse myeloma line is fused with a human B-lymphocyte
to
obtain a non-antibody-producing xenogeneic hybrid cell, such as the SPAZ-4
cell line.
The xenogeneic cell is then fused with an immunized human B-lymphocyte to
obtain an
antibody-producing trioma cell line. Triomas have been found to produce
antibodies more
stably than ordinary hybridomas made from human cells.
The B-lymphocytes are obtained from the blood, spleen, lymph nodes, or bone
marrow of a human donor. In vivo immunization of a living human with protein
is usually
undesirable because of the risk of initiating a harmful response. Thus, B-
lymphocytes are
usually immunized in vitro with a protein (e.g., MN) or an antigenic fragment
thereof, or a
cell bearing said protein (e.g., MN). Specific epitopic fragments consisting
essentially of
27

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
the amino acid segments that bind to one of the exemplified murine antibodies
may be
used for in vitro immunization. B-lymphocytes are typically exposed to antigen
for a
period of 7-14 days in a media such as RPMI-1640 (see, e.g., U.S. Patent No.
4,634,666)
supplemented with 10% human serum:
.5 The immunized B-lymphocytes may be fused= to a xenogeneic hybrid cell such
as
SPAZ-4 by methods known in the art. For example, the cells may be treated with
40-50%
polyethylene glycol of MW 1,000-4,000 for about 5-10 minutes at about 37 C.
Cells may
be separated from the fusion mixture and propagated in media selective for the
desired
hybrids (e.g., HAT or AH). Clones secreting antibodies having the required
binding
specificity may be identified by assaying the trioma culture medium for the
ability to bind
to a protein (e.g., MN) using the same methods as discussed above for non-
human
antibodies. Triomas producing human antibodies having the desired specificity
may be
sub-cloned, for example, by the limiting dilution technique and grown in vitro
in culture
medium.
Although triomas are genetically stable, they may not produce antibodies at
very
high levels. Expression levels may be increased by cloning antibody genes from
the
trioma into one or more expression vectors, and transforming the vector into a
cell line
such as the cell lines for expression of recombinant or humanized
immunoglobulins.
Human antibodies cross-reactive with a protein (e.g., MN) may also be produced
from non-human transgenic mammals having transgenes encoding at least a
segment of
the human immunoglobulin locus. Usually, the endogenous immunoglobulin locus
of
such transgenic mammals is functionally inactivated. The segment of the human
immunoglobulin locus may include unrearranged sequences of heavy and light
chain
components. Both inactivation of endogenous immunoglobulin genes and
introduction of
exogenous immunoglobulin genes may be achieved by targeted homologous
28

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
recombination, or by introduction of YAC chromosomes. The transgenic mammals
resulting from this process are capable of functionally rearranging the
immunoglobulin
component sequences, and expressing a repertoire of antibodies of various
isotypes
encoded by human immunoglobulin genes, without expressing endogenous
..5 inununoglobulin genes.= The production and properties of mammals having
these
properties are described in detail, for example, by Lonberg, et al., (WO
93/12227); and
Kucherlapati, (WO 91/10741) (each of which is incorporated by reference in its
entirety).
Cross-reacting MN human antibodies may be obtained by immunizing a transgenic
non-
human mammal as described above. Monoclonal antibodies may be prepared, for
example, by fusing B-cells from such mammals to suitable myeloma cell lines
using
conventional Kohler-Milstein technology (Kohler and Milstein, Nature 256:495-
497,
1975, incorporated by reference).
Mouse or other non-human antibodies that are cross-reactive with a protein
(e.g.,
MN) may be obtained using a variety of immunization strategies. In some
strategies, non-
human animals (usually non-human mammals such as mice) may be immunized with
MN
antigens. Immunogens may include cells stably transfected with MN and
expressing MN
on their cell surface, and MN protein or epitopic fragments containing the
segments of
these molecules that bind to the exemplified cross-reacting antibodies.
Antibody-
producing cells obtained from the immunized animals may be immortalized and
selected
for the production of an antibody which specifically binds to MN (e.g., Harlow
& Lane,
Antibodies, A Laboratory Manual, C.S.H.P., N.Y., 1988, incorporated by
reference).
Binding may be detected, for example, using an appropriate secondary antibody
bearing a
second label. Cross-reacting antibodies may then be further screened for their
capacity to
direct selective cellular cytotoxicity to cells expressing MN.
29

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
The present invention also relates to humanized antibodies having similar
binding
specificity and affinity to selected mouse or other non-human antibodies.
Humanized
antibodies may be formed by linking CDR regions (e.g., CDRI, CDR2, and CDR3)
of
non-human antibodies to a human framework and constant regions by recombinant
DNA
techniques (e.g., Queen, et al.,Troc. Natl. Acad. Sci. USA 86:10029-
10033,1989; WO
90/07861; each incorporated by reference in their entirety), i.e. CDR
grafting. These
humanized immunoglobulins have variable region framework residues
substantially from
a human immunoglobulin (referred to as an acceptor immunoglobulin) and
complementarity determining regions (CDRs) substantially from a mouse
immunoglobulin
(referred to as a donor immunoglobulin). The constant region(s), if present,
may also be
substantially from a human immunoglobulin.
In principal, a framework sequence from any human antibody may serve as the
template for CDR grafting. However, it has been demonstrated that straight CDR
replacement onto such a framework often leads to significant loss of binding
affinity to the
antigen (Glaser, et al., J. Immunol. 149:2606, 1992); Tempest, et al.,
Biotechnology 9:266,
1992; Shalaby, et al., J. Exp. Med. 17: 217, 1992). The more homologous a
human
antibody is to the original murine antibody, the less likely combining the
murine CDRs
with the human framework will be to introducing distortions into the CDRs that
could
reduce affinity. Therefore, homology (i.e, percent sequence identity) between
the
humanized antibody variable region framework and the donor antibody variable
region
framework of preferably at least about 65%, more preferably at least about
75%, still more
preferably at least about 85%, and yet more preferably about 95% or about 99%
is
suggested.
The heavy and light chain variable region framework residues may be derived
from the same or different human antibody sequences. However, heavy chain and
light

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
chain framework sequences chosen from the same human antibody reduce the
possibility
of incompatibility in assembly of the two chains. The human antibody sequences
may be
the sequences of naturally occurring human antibodies or may be consensus
sequences of
several human antibodies (e.g., WO 92/22653, incorporated by reference).
Certain amino
acids from the human variable region framework residues may=be selected for
substitution
based on their possible influence on CDR conformation and/or binding to
antigen.
Analysis of such possible influences may be accomplished by modeling,
examination of
the characteristics of the amino acids at particular locations, or empirical
observation of
the effects of substitution or mutagenesis of particular amino acids.
For example, when an amino acid differs between a murine variable region
framework residue and a selected human variable region framework residue, the
human
framework amino acid may be substituted by the equivalent framework amino acid
from
the mouse antibody when it is reasonably expected that the amino acid:
(1) contacts antigen directly,
(2) is adjacent to a CDR region in the sequence, or
(3) otherwise interacts with a CDR region (e.g., is within about 4-6 A of a
CDR
region).
Other candidates for substitution are, for example, acceptor human framework
amino acids that are unusual for a human immunoglobulin at that position.
These amino
acids may be substituted with amino acids from the equivalent position of the
donor
antibody or from the equivalent positions of more typical human
immunoglobulins. The
variable region frameworks of humanized immunoglobulins may show, for example,
at
least preferably about 85% sequence identity, more preferably at least about
90%
sequence identity, still more preferably at least about 95% sequence identity,
and even
31

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
more preferably at least about 99% sequence identity, to a human variable
region
framework sequence or consensus of such sequences.
The present invention also relates to bispecific or bifunctional antibodies
that have
one binding site that specifically binds to a protein (e.g., MN) and a second
binding site
==5 that specifically binds to=a second moiety-. In bi-specific antibodies,
one.heavy and light
chain pair is usually from, for example, an MN binding antibody and the other
pair from
an antibody raised against another epitope. This results in multi-functional
valency, that
is, an ability to bind at least two different epitopes simultaneously.
Binding Assays
Any useful means to describe the strength of binding (or affinity) between an
antibody or antibody fragment of the invention and an antigen of the invention
(MN
protein) can be used. For example, the dissociation constant, Kd (Kd = k2/kl =
[antibody] [antigen] / [antibody-antigen complex]) can be determined by
standard kinetic
analyses that are known in the art. It will be appreciated by those of
ordinary skill in the
art that the dissociation constant indicates the strength of binding between
an antibody and
an antigen in terms of how easy it is to separate the complex. If a high
concentration of
antibody and antigen are required to form the complex, the strength or
affinity of binding
is low, resulting in a higher Kd. It follows that the smaller the Kd (as
expressed in
concentration units, e.g. molar or nanomolar), the stronger the binding.
Affinity can be assessed and/or measured by a variety of known techniques and
immunoassays, including, for example, enzyme-linked immunospecific assay
(ELISA),
Bimolecular Interaction Analysis (BIA) (e.g., Sjolander and Urbaniczky, Anal.
Chem.
63:2338-2345, 1991; Szabo, et al_, Curr. Opin. Struct. Biol. 5:699-705, 1995,
each
incorporated herein by reference), and fluorescence-activated cell sorting
(FACS) for
quantification of antibody binding to cells that express MN. BIA is a
technology for
32

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
analyzing biospecific interactions in real time, without labeling any of the
interactants
(e.g., BlAcoreTM). BlAcore is based on determining changes in the optical
phenomenon
surface plasmon resonance (SPR) in real-time reactions between biological
molecules,
such as, an antibody of the invention and an MN protein antigen. References
relating to
.=5 BlAcore technology can-be further found in U.S. Published Application Nos:
2006/0223113, 2006/0134800, 2006/0094060, 2006/0072 1 1 5, 2006/0019313,
2006/0014232, and 2005/0199076, each of which are incorporated herein in their
entireties
by reference.
Antibodies or antigen binding antibody fragments of the invention that
specifically
bind to a protein (e.g., MN) provide a detection signal, for example,
preferably at least
about 5-fold higher, more preferably at least about 10-fold higher, and still
more
preferably at least about 20-fold higher than a detection signal provided for
other proteins
when used in an immunochemical assay_ As such, these antibodies may be used to
immunoprecipitate a protein (e.g., MN) from solution.
The antibodies and fragments of the invention may be assayed for
immunospecific
binding (or binding that is "specifically immunoreactive," which is herein
defined) by any
suitable method known in the art. Assays involving an antibody and an antigen
are known
as "irnmunoassays," which can be employed in the present invention to
characterize both
the antibodies and the antigens of the invention. The immunoassays which can
be used
include but are not limited to competitive and non-competitive assay systems
using
techniques such as western blots, radioimmunoassays, ELISA (enzyme linked
immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays,
precipitin
reactions, gel diffusion precipitin reactions, immunodiffusion assays,
agglutination assays,
complement-fixation assays, immunoradiometric assays, fluorescent
immunoassays,
protein A immunoassays, to name but a few. Such assays are routine and well
known in
33

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
the art (see, e.g., Ausubel et al., eds, 1994, Current Protocols in Molecular
Biology, Vol. 1,
John Wiley & Sons, Inc., New York, which is incorporated by reference herein
in its
entirety) and can be performed without undue experimentation. Exemplary
immunoassays
are described briefly below (but are not intended by way of limitation).
Irnmunoprecipitation protocols- generally comprise lysing a population of
cells in a
lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium
deoxycholate,
0.1% SDS, 0.15 M NaCI, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol)
supplemented
with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF,
aprotinin, sodium
vanadate), adding the antibody of interest to the cell lysate, incubating for
a period of time
(e.g., 14 hours) at 4 C, adding protein A and/or protein G sepharose beads to
the cell
lysate, incubating for about an hour or more at 4 C, washing the beads in
lysis buffer and
resuspending the beads in SDS/sample buffer. The ability of the antibody of
interest to
immunoprecipitate a particular antigen can be assessed by, e.g., western blot
analysis.
One of skill in the art would be knowledgeable as to the parameters that can
be modified
to increase the binding of the antibody to an antigen and decrease the
background (e.g.,
pre-clearing the cell lysate with sepharose beads). For further discussion
regarding
immunoprecipitation protocols see, e.g., Ausubel et al., eds, 1994, Current
Protocols in
Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1, which
is
incorporated herein by reference.
Western blot analysis generally comprises preparing protein samples,
electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8% 20%
SDS-PAGE
depending on the molecular weight of the antigen), transferring the protein
sample from
the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon,
blocking the
membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing
the
membrane in washing buffer (e.g., PBS-Tween 20), blocking the membrane with
primary
34

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
antibody (the antibody of interest) diluted in blocking buffer, washing the
membrane in
washing buffer; blocking the membrane with a secondary antibody (which
recognizes the
primary antibody, e.g., an anti-human antibody) conjugated to an enzymatic
substrate
(e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule
(e.g., 32P or
--5 1251) diluted in blocking buffer, washing the -membrane =in wash- buffer,
and detecting the
presence of the antigen. One of skill in the art would be knowledgeable as to
the
parameters that can be modified to increase the signal detected and to reduce
the
background noise. For further discussion regarding western blot protocols see,
e.g.,
Ausubel et al., eds, 1994, Current Protocols in Molecular Biology, Vol. 1,
John Wiley &
Sons, Inc., New York at 10.8.1, which is incorporated herein by reference.
ELISAs typically comprise preparing antigen, coating the well of a 96 well
microtiter plate with the antigen, adding the antibody of interest conjugated
to a detectable
compound such as an enzymatic substrate (e.g., horseradish peroxidase or
alkaline
phosphatase) to the well and incubating for a period of time, and detecting
the presence of
the antigen. In ELISAs the antibody of interest does not have to be conjugated
to a
detectable compound; instead, a second antibody (which recognizes the antibody
of
interest) conjugated to a detectable compound may be added to the well.
Further, instead
of coating the well with the antigen, the antibody may be coated to the well.
In this case, a
second antibody conjugated to a detectable compound may be added following the
addition of the antigen of interest to the coated well. One of skill in the
art would be
knowledgeable as to the parameters that can be modified to increase the signal
detected as
well as other variations of ELISAs known in the art. For further discussion
regarding
ELISAs see, e.g., Ausubel et al., eds, 1994, Current Protocols in Molecular
Biology, Vol.
1, John Wiley & Sons, Inc., New York at 11.2.1, which is incorporated herein
by
reference.

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
In one aspect, the present invention embodies a number of different antibodies
having MN binding characteristics identified by screening the MorphoSys HuCAL
GOLD
Fab library. In one aspect of the invention, the amino acid sequences of three
CDRs of the
VH region of a human antibody were identified (SEQ ID NOS: 57-85; Figure 2).
In
another embodiment=of the invention, the amino -acid sequences of three-CDRs
of the VL
region of a human MN antibody were identified (SEQ ID NOS: 86-112; Figure 2).
The
present invention also relates to combinations of CDRs, frameworks, and VH/VL
pairs.
Examples of such combinations are shown in Figure 3 (SEQ ID NOS: 113-132 for
the
encoding nucleotide sequences) and in Figure 4 )SEQ ID NOS 133-152 for the
protein
sequences). Antibodies that have MN binding are also shown in Figure 4.
Details of the
screening process are described in the examples described herein. Other
selection
methods for highly active specific antibodies or antibody fragments may be
envisioned by
those skilled in the art and used to identify human MN antibodies.
Antibodies and/or antigen binding antibody fragments of the invention may also
be
purified from any cell that expresses the antibodies, including host cells
that have been
transfected with antibody-encoding expression constructs. The host cells may
be cultured
under conditions whereby the antibodies are expressed. Purified antibodies
and/or antigen
binding antibody fragments may be separated from other cellular components
that may
associate with the antibodies in the cell, such as certain proteins,
carbohydrates, or lipids
using methods well known in the art. Such methods include, but are not limited
to, size
exclusion chromatography, ammonium sulfate fractionation, ion exchange
chromatography, affinity chromatography, and preparative gel electrophoresis.
Purity of
the preparations may be assessed by any means known in the art, such as SDS-
polyacrylamide gel electrophoresis. A preparation of purified antibodies may
contain
36

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
more than one type of antibody (e.g., antibodies with the MN binding and
neutralizing
characteristics).
Alternatively, antibodies may be produced using chemical methods to synthesize
its amino acid sequence or portions of the antibody sequence (e.g. CDR
sequences), such
as by direct peptide- synthesis using solid-phase techniques (e.g.;
Merrifield, J. Am. Chem.
Soc. 85:2149-2154, 1963; Roberge, et al., Science 269:202-204, 1995, each of
which are
incorporated herein by reference). Protein synthesis may be performed using
manual
techniques or by automation. Automated synthesis may be achieved, for example,
using
Applied Biosystems 431A Peptide Synthesizer (Perkin Elmer). Optionally,
fragments of
antibodies may be separately synthesized and combined using chemical methods
to
produce a full-length molecule.
Once an antibody or fragment in accordance with the invention is identified or
obtained, for example, by any of the methods herein described, for example,
including by
traditional methods of antibody production (e.g. animal immunization methods),
monoclonal production, or by recombinant DNA means, it may be preferable to
carry out
further steps to characterize and/or purify and/or modify the antibody. For
example, it
may be desirable to prepare an immunoreactive antibody fragment or to prepare
a purified,
high-titer composition of the identified, desirable antibody or to test the
immunoreactivity
of the identified antibody or fragment thereof. The present invention
contemplates any
known and suitable methods for characterizing, purifying, or assaying the
antibodies of the
present invention and it is expected the any person of ordinary skill in the
art to which the
invention pertains will have the requisite level of technical know-how and
resources, e.g.
technical manuals or treatises, to accomplish any further characterization,
purification
and/or assaying of the antibodies of the invention without undue
experimentation.
37

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
In particular aspects, the antibodies and/or antibody fragments of the
invention can
be recovered and purified from recombinant cell cultures by well-known methods
including, but not limited to, protein A purification, ammonium sulfate or
ethanol
precipitation, acid extraction, anion or cation exchange chromatography,
phosphocellulose
chromatography, hydrophobic interaction chromatography, affinity
chromatography,
hydroxylapatite chromatography and lectin chromatography. High performance
liquid
chromatography ("HPLC") can also be employed for purification_ See, e.g.,
Colligan,
Current Protocols in Immunology, or Current Protocols in Protein Science, John
Wiley &
Sons, NY, N.Y., (1997 2001), e.g., Chapters 1, 4, 6, 8, 9, 10, each entirely
incorporated
herein by reference.
The term "isolating" in the context of antibodies refers to any process known
in the
art for purifying antibodies. Methods for purifying antibodies are well known
in the art
and the present invention contemplates any suitable method that would be known
to the
skilled person and which would not require undue experimentation. For example,
chromatographic methods, such as, for example, immuno-affinity chromatography
(immobilized ligand to bind and trap antibody of interest), affinity
chromatography,
protein precipitation, ion exchange chromatography, hydrophobic interaction
chromatography, size-exclusion chromatography, as well as electrophoresis, can
be found
described in the technical literature, for example, in Methods in Enzymology,
Volume
182, Guide to Protein Purification, Eds. J. Abelson, M. Simon, Academic Press,
ls'
Edition, 1990, which is incorporated herein by reference. Thus, suitable
materials for
performing such purification steps, such as chromatographic steps, are known
to those
skilled in the art. Such methods are suitable for purification of any of the
antibodies,
antigens or any fragments thereof that are in accordance with the invention as
described
herein.
38

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Certain embodiments may require the purification or isolation of expressed
proteins or antibodies or fragments thereof from a host cell or a portion
thereof.
Conventional procedures for isolating recombinant proteins from transformed
host cells
are contemplated by the present invention. Such methods include, for example,
isolation
of the protein or fragments-of interest by initial extraction from cell
pellets or from cell
culture medium, followed by salting-out, and one or more chromatography steps,
including aqueous ion exchange chromatography, size exclusion chromatography
steps,
high performance liquid chromatography (HPLC), and affinity chromatography may
be
used to isolate the recombinant protein or fragment. Guidance in the
procedures for
protein purification can be found in the technical literature, including, for
example,
Methods in Enzymology, Volume 182, Guide to Protein Purification, Eds. J.
Abelson, M.
Simon, Academic Press, lst Edition, 1990, which is already incorporated by
reference.
Chemically-synthesized molecules may be substantially purified by preparative
high performance liquid chromatography (see, e.g., Creighton, Proteins:
Structures and
Molecular Principles, WH Freeman and Co., New York, N.Y., 1983, incorporated
herein
by reference). The composition of a synthetic polypeptide may be confirmed by
amino
acid analysis or sequencing (e.g., using Edman degradation).
Potynucleotides
In another aspect, the present invention relates to polynucleotides encoding
the
antibodies (e.g., antibodies against MN) or the antigen binding antibody
fragments of the
invention. These polynucleotides may be used, for example, to produce
quantities of the
antibodies for therapeutic or diagnostic use or the produce samples of
antibodies or
fragments for use in imxnunoassays of the invention.
Polynucleotides that may be used to encode, for example, each of three CDRs
within antibody VH regions are described by SEQ IIa NOS: 1-29.
Polyniucleotides that
39

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
may be used to encode, for example, each of three CDRs within antibody VL
regions are
described by SEQ ID NOS: 30-56. Polynucleotides that encode, for example,
complete
heavy chain and light chain variable regions of antibodies are described by
SEQ ID NOS:
113-132 (Figure 3).
Polynucleotides of the present invention may also be isolated from host cells,
free
of other cellular components such as membrane components, proteins, and lipids
according to any known or suitable method in the art. Polynucleotides may be
isolated
using standard nucleic acid purification techniques, or synthesized using an
amplification
technique such as the polymerase chain reaction (PCR), or by using an
automatic
synthesizer. Methods for isolating polynucleotides are routine and are known
in the art.
Any such technique for obtaining a polynucleotide may be used to obtain
isolated
polynucleotides encoding antibodies of the invention. For example, restriction
enzymes
and probes may be used to isolate polynucleotides which encode antibodies.
Antibody-encoding cDNA molecules may be made with standard molecular
biology techniques, using mRNA as a template. Thereafter, cDNA molecules may
be
replicated using molecular biology techniques known in the art and disclosed
in manuals
such as Sambrook, et al., (Molecular Cloning: A Laboratory Manual, (Second
Edition,
Cold Spring Harbor Laboratory Press; Cold Spring Harbor, N.Y.; 1989, Vol. 1-3,
incorporated herein by reference). An amplification technique, such as PCR,
may be used
to obtain additional copies of the polynucleotides.
Alternatively, synthetic chemistry techniques may be used to synthesize
polynucleotides encoding antibodies of the invention. The degeneracy of the
genetic code
allows alternate nucleotide sequences to be synthesized that will encode an
antibody
having, for example, one of the VH-CDR1, VH-CDR2, VH-CDR3, VL-CDR1, VL-

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
CDR2, VL-CDR3, complete VH or complete VL amino acid sequences (e.g., SEQ ID
NOS: 57-112 and 133-152).
To express a polynucleotide encoding an antibody, the polynucleotide may be
inserted into an expression vector that contains the necessary elements for
the transcription
.5 and translation of the inserted coding sequence. Methods that are well
known to those
skilled in the art may be used to construct expression vectors containing
sequences
encoding antibodies and appropriate transcriptional and translational control
elements.
These methods include in vitro recombinant DNA techniques, synthetic
techniques, and in
vivo genetic recombination. Such techniques are described, for example, in
Sambrook, et
al. (1989) and in Ausubel, et al., (Current Protocols in Molecular Biology,
John Wiley &
Sons, New York, N.Y., 1995, incorporated herein by reference).
A variety of expression vector/host systems may be utilized to contain and
express
sequences encoding antibodies. These include, but are not limited to,
m.icroorganisms,
such as bacteria transformed with recombinant bacteriophage, plasmid, or
cosmid DNA
expression vectors; yeast transformed with yeast expression vectors; insect
cell systems
infected with virus expression vectors (e.g., baculovirus); plant cell systems
transformed
with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco
mosaic
virus, TMV); or bacterial expression vectors (e.g., Ti or pBR322 plasmids), or
animal cell
systems.
The control elements or regulatory sequences are those non-translated regions
of
the vector -- enhancers, promoters, 5' and 3' untranslated regions -- which
interact with
host cellular proteins to carry out transcription and translation. Such
elements may vary in
strength and specificity_ Depending on the vector system and host utilized,
any number of
suitable transcription and translation elements, including constitutive and
inducible
promoters, may be used. For example, when cloning in bacterial systems,
inducible
41

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
promoters such as the hybrid lacZ promoter of the BLUESCRIPT phagemid
(Stratagene,
LaJolla, Calif.), pSPORTl plasmid (Life Technologies), or the like can be
used. The
baculovirus polyhedrin promoter may be used in insect cells. Promoters or
enhancers
derived from the genomes of plant cells (e.g., heat shock, RUBISCO, and
storage protein
=5 genes) or from plant viruses (e.g., viral promoters or leader sequences)
may be cloned into
the vector. In mammalian cell systems, promoters from mammalian genes or from
mammalian viruses may be used, e.g. a CMV promoter. If it is necessary to
generate a
cell line that contains multiple copies of a nucleotide sequence encoding an
antibody,
vectors based on SV40 or EBV may be used with an appropriate selectable
marker.
Accordingly, the present invention also relates to recombinant vectors that
include
isolated nucleic acid molecules of the present invention (e.g. the heavy and
light chain
variable regions of SEQ ID NOS: 113-132), host cells that are genetically
engineered with
the recombinant vectors, and the production and/or expression of the
recombinant
antibodies or fragments thereof of the invention.
The expression constructs can further contain sites for transcription
initiation,
termination and, in the transcribed region, a ribosome binding site for
translation. The
coding portion of the mature transcripts expressed by the constructs can
further include a
translation initiating at the beginning and a termination codon (e.g., UAA,
UGA or UAG)
appropriately positioned at the end of the mRNA to be translated, with UAA and
UAG
preferred for mammalian or eukaryotic cell expression.
Expression vectors can also include at least one selectable marker. Such
markers
include, but are not limited to, mammalian cell markers, such as, methotrexate
(MTX),
dihydrofolate reductase (DHFR) (see e.g., U.S. Patents Nos. 4,399,216;
4,634,665;
4,656,134; 4,956,288; 5,149,636; and 5,179,017, each of which are incoporated
by
reference), ampicillin, neomycin (G418), mycophenolic acid, or glutamine
synthetase
42

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
(GS) (see e.g., U.S. Pat. Nos. 5,122,464; 5,770,359; 5,827,739, each of which
are
incorporated by reference), and bacterial cell markers, such as, tetracycline
or ampicillin
resistance genes. Appropriate culture mediums and conditions for the above-
described
host cells are known in the art. Suitable vectors will be readily apparent to
the skilled
= 5 artisan. = = - = = -
Any suitable known method for introducing a DNA of the invention, e.g. a DNA
expression vector containing one or more antibody-encoding sequences of SEQ ID
NOS:
113-132, into a host cell can be utilized. Some known methods include calcium
phosphate
transfection, DEAE-dextran mediated transfection, cationic lipid-mediated
transfection,
electroporation, transduction, infection or other known methods. Such methods
are
described in the art, such as in Sambrook, et al., (Molecular Cloning: A
Laboratory
Manual, (Second Edition, Cold Spring Harbor Laboratory Press; Cold Spring
Harbor,
N.Y.; 1989) Vol. 1-3, Chapters 14, 16 and 18).
Those of ordinary skill in the art are knowledgeable in the numerous
expression
systems available for expression of a nucleic acid encoding the antibodies or
portions
thereof of the present invention.
Illustrative of cell cultures useful for the production of the antibodies or
antibody
fragments of the invention are mammalian cells. Mammalian cell systems often
will be in
the form of monolayers of cells although mammalian cell suspensions or
bioreactors can
also be used. A number of suitable host cell lines capable of expressing
intact glycosylated
proteins have been developed in the art, and include the CHO, CHO-S, COS-1
(e.g.,
ATCC CRL 1650), COS-7 (e.g., ATCC CRL-1651), HEK293, BHK21 (e.g., ATCC CRL-
10), CHO (e.g., ATCC CRL 1610) and BSC-l (e.g., ATCC CRL-26) cell lines, Cos-7
cells, CHO cells, hep G2 cells, P3X63Ag8.653, SP2/0-Ag14, 293 cells, HeLa
cells and the
like, which are readily available from, for example, American Type Culture
Collection.
43

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Expression vectors for these cells can include one or more of the following
expression control sequences, such as, but not limited to an origin of
replication; a
promoter (e.g., late or early SV40 promoters, the CMV promoter (e.g., U.S.
Pat. Nos.
5,168,062; 5,385,839), an HSV tk promoter, a pgk (phosphoglycerate kinase)
promoter, an
EF-1 alpha promoter (U.S. Pat: No. 5,266,491); an immunoglobulin promoter; an
enhancer, and/or processing information sites, such as ribosome binding sites,
RNA splice
sites, polyadenylation sites (e.g., an SV40 large T Ag poly A addition site),
and
transcriptional terminator sequences. Other cells useful for production of
nucleic acids or
proteins of the present invention are known and/or available, for instance,
from the
American Type Culture Collection Catalogue of Cell Lines and Hybridomas or
other
known or commercial sources.
When eukaryotic host cells are employed, polyadenlyation or transcription
terminator sequences can be incorporated into the vector. An example of a
terminator
sequence is the polyadenlyation sequence from the bovine growth hormone gene.
] 5 Sequences for accurate splicing of transcripts can also be included. An
example of a
splicing sequence is the VPI intron from SV40 (Sprague, et al., J. Virol.
45:773 781
(1983)). Additionally, gene sequences to control replication in the host cell
can be
incorporated into the vector, as known in the art.
General texts describing additional molecular biological techniques useful
herein,
including the preparation of antibodies include Berger and Kimmel (Guide to
Molecular
Cloning Techniques, Methods in Enzymology, Vol. 152, Academic Press, Inc.);
Sambrook, et al., (Molecular Cloning: A Laboratory Manual, (Second Edition,
Cold
Spring Harbor Laboratory Press; Cold Spring Harbor, N.Y.; 1989) Vol. 1-3);
Current
Protocols in Molecular Biology, (F. M. Ausabel et al. [Eds.], Current
Protocols, a joint
venture between Green Publishing Associates, Inc. and John Wiley & Sons, Inc.
44

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
(supplemented through 2000)); Harlow et al., (Monoclonal Antibodies: A
Laboratory
Manual, Cold Spring Harbor Laboratory Press (1988), Paul [Ed.]); Fundamental
Immunology, (Lippincott Williams & Wilkins (1998)); and Harlow, et al., (Using
Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press (1998)),
all of
which are incorporated herein by reference.
In another embodiment, the present invention also relates to the use of
quantitative
imrnunoassays to measure levels of MN protein in patient samples. Many formats
may be
adapted for use with the methods of the present invention. For example, the
detection and
quantitation of MN protein in patient samples may be performed, by enzyme-
linked
immunosorbent assays, radioimmunoassays, dual antibody sandwich assays,
agglutination
assays, fluorescent immunoassays, immunoelectron and scanning microscopy,
among
other assays commonly known in the art. The quantitation of MN protein in such
assays
may be adapted by conventional methods known in the art. In one embodiment,
serial
changes in circulating MN protein levels may be detected and quantified by a
sandwich
assay in which the capture antibody has been immobilized using conventional
techniques
on the surface of the support.
Suitable supports include, for example, synthetic polymer supports, such as
polypropylene, polystyrene, substituted polystyrene, polyacrylamides (such as
polyamides
and polyvinylchloride), glass beads, agarose, and nitrocellulose.
An example of an ELISA sandwich immunoassay that may be used in the methods
of the present invention, uses mouse anti-human MNN monoclonal antibody as the
capture
antibody and biotinylated goat anti-human MN polyclonal antibody as the
detector
antibody. The capture monoclonal antibody is immobilized on microtiter plate
wells.
Diluted human serum/plasma samples or MN standards (recombinant wild-type MN
protein) are incubated in the wells to allow binding of MN antigen by the
capture

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
monoclonal antibody. After washing of wells, the immobilized MN antigen is
exposed to
a biotinylated detector antibody after which the wells are again washed. A
streptavidin-
horseradish peroxidase conjugate is then added. After a final wash, TMB Blue
Substrate
is added to the wells to detect bound peroxidase activity. The reaction is
stopped by the
addition of 2.5 N sulfuric acid, and the absorbance is measured at 450 nm.
Correlating the
absorbance values of samples with the MN standards allows the determination of
a
quantitative value of MN in pg/ml of serum or plasma.
The antibodies useful to identify MN proteins may be labeled in any
conventional
manner. An example of a label is horseradish peroxidase, and an example of a
method of
labeling antibodies is by using biotin-strepavidin complexes.
As appropriate, antibodies used in the immunoassays of this invention that are
used
as tracers may be labeled in any manner, directly or indirectly, that results
in a signal that
is visible or can be rendered visible. Detectable marker substances include
radionuclides,
such as 3H, 1251 and 13 11; fluorescers, such as, fluorescein isothiocyanate
and other
fluorochromes, phycobiliproteins, phycoerythin, rare earth chelates, Texas
red, dansyl and
rhodamine; colorimetric reagents (chromogens); electron-opaque materials, such
as
colloidal gold; bioluminescers; chemiluminescers; dyes; enzymes, such as,
horseradish
peroxidase, alkaline phosphatases, glucose oxidase, glucose-6-phosphate
dehydrogenase,
acetylcholinesterase, alpha -, beta-galactosidase, among others; coenzymes;
enzyme
substrates; enzyme cofactors; enzyme inhibitors; enzyme subunits; metal ions;
free
radicals; or any other immunologically active or inert substance which
provides a means
of detecting or measuring the presence or amount of immunocomplex formed.
Exemplary
of enzyme substrate combinations are horseradish peroxidase and tetramethyl
benzidine
(TMB), and alkaline phosphatases and paranitrophenyl phosphate (pNPP).
46

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Another detection and quantitation systems according to this invention produce
luminescent signals, bioluminescent (BL) or chemiluminescent (CL). In
chemiluminescent (CL) or bioluminescent (BL) assays, the intensity or the
total light
emission is measured and related to the concentration of the unknown analyte.
Light can
be measured quantitatively using a Iuminometer (.photomultiplier tube as the
detector) or
charge-coupled device, or qualitatively by means of photographic or X-ray
film. The main
advantages of using such assays is their simplicity and analytical
sensitivity, enabling the
detection and/or quantitation of very small amounts of analyte.
Exemplary luminescent labels are acridinium esters, acridinium sulfonyl
carboxarnides, luminol, umbelliferone, isoluminol derivatives, photoproteins,
such as
aequorin, and luciferases from fireflies, marine bacteria, Vargulla and
Renilla. Luminol
can be used optionally with an enhancer molecule such as 4-iodophenol or
4-hydroxy-cinnamic acid. Typically, a CL signal is generated by treatment with
an
oxidant under basic conditions.
Additional luminescent detection systems are those wherein the signal
(detectable
marker) is produced by an enzymatic reaction upon a substrate. CL and BL
detection
schemes have been developed for assaying alkaline phosphatases (AP), glucose
oxidase,
glucose 6-phosphate dehydrogenase, horseradish peroxidase (HRP), and xanthine-
oxidase
labels, among others. AP and HRP are two enzyme labels which can be
quantitated by a
range of CL and BL reactions. For example, AP can be used with a substrate,
such as an
adamantyl 1,2-dioxetane aryl phosphate substrate (e.g. AMPPD or CSPD; Kricka,
L.J.,
"Chemiluminescence and Bioluminescence, Analysis by," Molecular Biology and
Biotechnology: A Comprehensive Desk Reference (ed. R.A. Meyers) (VCH
Publishers;
N.Y., N.Y.; 1995)); for example, a disodium salt of 4-methoxy-4-(3-
phosphatephenyl)
spiro [1,2-dioxetane-3,2'-adamantane], with or without an enhancer molecule
such as
47

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
1-(trioctylphosphonium methyl)-4- (tributylphosphonium methyl) benzene
diochloride.
HRP is may be used with substrates, such as, 2',3',6'-trifluorophenyl-methoxy-
10-
methylacridan-9-carboxylate.
CL and BL reactions may be adapted for analysis not only of enzymes, but also
of
=.5 other substrates, cofactors, inhibitors, metal ions, and the like. For
example, luminol,
firefly luciferase, and marine bacterial luciferase reactions are indicator
reactions for the
production or consumption of peroxide, ATP, and NADPH, respectively. They may
be
coupled to other reactions involving oxidases, kinases, and dehydrogenases,
and may be
used to measure any component of the coupled reaction (enzyme, substrate,
cofactor).
The detectable marker may be directly or indirectly linked to an antibody used
in
an assay of this invention. Exemplary of an indirect linkage of the detectable
label is the
use of a binding pair between an antibody and a marker or the use of a signal
amplification
system.
Examples of binding pairs that may be used to link antibodies to detectable
markers are biotin/avidin, streptavidin, or anti-biotin; avidin/anti-avidin;
thyroxine/thyroxine-binding globulin; antigen/antibody; antibody/ anti-
antibody;
carbohydrate/lectins; hapten/anti-hapten antibody; dyes and hydrophobic
molecules/hydrophobic protein binding sites; enzyme inhibitor, coenzyme or
cofactor/enzyme; polynucleic acid/homologous polynucleic acid sequence;
fluorescein/anti- fluorescein; dinitrophenol/anti-dinitrophenol; vitamin B
12/intrinsic
factor; cortisone, cortisol/cortisol binding protein; and ligands for specific
receptor
protein/membrane associated specific receptor proteins.
Various means for linking labels directly or indirectly to antibodies are
known in
the art. For example, labels may be bound either covalently or non-covalently.
Exemplary antibody conjugation methods are described in Avarmeas, et al.,
Scan. J.
48

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Immunol. 8(Suppl. 7): 7, 1978); Bayer, et al., Meth. Enzymol. 62:308, 1979;
Chandler, et
al., J. Immunol. Meth. 53:187, 1982; Ekeke and Abuknesha, J. Steroid Biochem.
11: 1579,
1979; Engvall and Perlmann, J. Irnmunol. 109:129, 1972; Geoghegan, et al.,
Immunol.
Comm. 7:1, 1978; and Wilson and Nakane, Immunofluorescence and Related
Techniques,
Elsevier/North Holland Biomedical Press; Amsterdam (1978), each of which are
incorporate herein by reference.
Depending upon the nature of the label, various techniques may be employed for
detecting and quantitating the label. For fluorescers, a large number of
fluorometers are
available. For chemiluminescers, luminometers or films are available. With
enzymes, a
fluorescent, chemiluminescent, or colored product may be determined or
measured
fluorometrically, luminometrically, spectrophotometrically, or visually.
Various types of chemiluminescent compounds having an acridinium,
benzacridinium, or acridan type of heterocyclic ring systems are other
examples of labels.
Examples of acridinium esters include those compounds having heterocyclic
rings or ring
systems that contain the heteroatom in a positive oxidation state including
such ring
systems as acridinium, benz[a]acridinium, benz[b]acridinium,
benz[c]acridinium, a
benzimidazole cation, quinolinium, isoquinolinium, quinolizinium, a cyclic
substituted
quinolinium, phenanthridinium, and quinoxalinium.
The tracer may be prepared by attaching to the selected antibody either
directly or
indirectly a reactive functional group present on the acridinium or
benzacridinium ester, as
is well known to those skilled in the art (see, e.g., Weeks, et al., Clin.
Chem. 29(8):1474-
1479, 1983). Examples of compounds are acridinium and benzacridinium esters
with an
aryl ring leaving group and the reactive functional group present in either
the para or the
meta position of the aryl ring. (e.g., U.S. Patent No. 4,745,181 and WO
94/21823,
incorporated herein by reference).
49

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Methods of Use
The term "treatment" includes any process, action, application, therapy, or
the like,
wherein a subject (or patient), including a human being, is provided medical
aid with the
object of improving the subject's condition, directly or indirectly, or
slowing the
progression of a-condition or disorder= in the subject, or ameliorating at
least one symptom
of the disease or disorder under treatment.
The term "combination therapy" or "co-therapy" means the administration of two
or more therapeutic agents to treat a disease, condition, and/or disorder.
Such
administration encompasses co-administration of two or more therapeutic agents
in a
substantially simultaneous manner, such as in a single capsule having a fixed
ratio of
active ingredients or in multiple, separate capsules for each inhibitor agent.
In addition,
such administration encompasses use of each type of therapeutic agent in a
sequential
manner. The order of administration of two or more sequentially co-
administered
therapeutic agents is not limited.
The phrase "therapeutically effective amount" means the amount of each agent
adininistered that will achieve the goal of improvement in a disease,
condition, and/or
disorder severity, and/or symptom thereof, while avoiding or minimizing
adverse side
effects associated with the given therapeutic treatment.
The term "pharmaceutically acceptable" means that the subject item is
appropriate
for use in a pharmaceutical product.
The antibodies of this invention are expected to be valuable as therapeutic
agents.
Accordingly, an embodiment of this invention includes a method of treating the
various
conditions in a patient (including mammals) which comprises administering to
said patient
a composition containing an amount of an antibody of the invention that is
effective in
treating the target condition.

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
The antibodies of the present invention may be used in the treatment or
prevention
of diseases and/or behaviors that are associated with the MN protein. These
diseases
and/or behaviors include, for example, cancer, such as, carcinomas of the
kidney,
esophagus, breast, cervix, colon, and lung. The present invention also relates
to methods
of ameliorating symptom-s of a disorder in which MN is elevated or otherwise
abnormally
expressed. These disorders include, without limitation, carcinomas of the
kidney,
esophagus, breast, cervix, colon, and lung (see, e.g., (Liao, Cancer Res.
57:2827-283 l,
1997; Turner, Hum. Pathol. 28:740-744, 1997; Liao, et al., Am. J. Pathol.
145:598-609,
1994; Saarnio, et al., Am. J. Pathol. 153:279-285, 1998; Vermylen, et al.,
Eur. Respir. J.
14:806-811, 1999). In one embodiment of the invention, a therapeutically
effective dose
of an antibody of the invention is administered to a patient having a disorder
in which MN
is elevated.
Antibodies of the present invention may be administered alone or in
combination
with one or more additional therapeutic agents. Combination therapy includes
administration of a single pharmaceutical dosage formulation which contains an
antibody
of the present invention and one or more additional therapeutic agents, as
well as
administration of the antibody of the present invention and each additional
therapeutic
agents in its own separate pharmaceutical dosage formulation. For example, an
antibody
of the present invention and a therapeutic agent may be administered to the
patient
together in a single oral dosage composition or each agent may be administered
in separate
oral dosage formulations.
Where separate dosage formulations are used, the antibody of the present
invention
and one or more additional therapeutic agents may be administered at
essentially the same
time (e.g., concurrently) or at separately staggered times (e.g.,
sequentially). The order of
administration of the agents is not limited.
51

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
For example, in one aspect, co-administration of an anti-MN antibody or
antibody
fragment of the invention together with one or more anti-cancer agents to
potentiate the
effect of either the antibody/fragment or the anti-cancer agent(s) or both is
contemplated
for use in treating MN-related disorders, such as, cancer.
Such combination-therapies -may also be used to prevent cancer, prevent the
recurrence of cancer, prevent the spread or metastasis of a cancer, or reduce
or ameliorate
the symptoms associated with cancer.
The one or more anti-cancer agents can include any known and suitable compound
in the art, such as, for example, chemoagents, other immunotherapeutics,
cancer vaccines,
anti-angiogenic agents, cytokines, hormone therapies, gene therapies, and
radiotherapies.
A chemoagent (or "anti-cancer agent" or "anti-tumor agent" or "cancer
therapeutic") refers
to any molecule or compound that assists in the treatment of a cancer.
Examples of
chemoagents contemplated by the present invention include, but are not limited
to,
cytosine arabinoside, taxoids (e.g., paclitaxel, docetaxel), anti-tubulin
agents (e.g.,
paclitaxel, docetaxel, epothilone B, or its analogues), macrolides (e.g.,
rhizoxin) cisplatin,
carboplatin, adriamycin, tenoposide, mitozantron, discodermolide,
eleutherobine, 2-
chlorodeoxyadenosine, alkylating agents (e.g., cyclophosphamide,
mechlorethamine,
thioepa, chlorambucil, melphalan, carmustine (BSNU), lomustine (CCNU),
cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and
cis-
dichlorodiamine platinum (lI) (DDP) cisplatin, thio-tepa), antibiotics (e.g.,
dactinomycin
(formerly actinomycin), bleomycin, mithramycin, anthramycin), antimetabolites
(e.g.,
methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, flavopiridol, 5-
fluorouracil,
fludarabine, gemcitabine, dacarbazine, temozolamide), asparaginase, Bacillus
Calmette
and Guerin, diphtheria toxin, hexamethylmelamine, hydroxyurea, LYSODREN®,
nucleoside analogues, plant alkaloids (e.g., Taxol, paclitaxel, camptothecin,
topotecan,
52

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
irinotecan (CAMPTOSAR, CPT-11), vincristine, vinca alkyloids such as
vinblastine),
podophyllotoxin (including derivatives such as epipodophyllotoxin, VP-16
(etoposide),
VM-26 (teniposide)), cytochalasin B, colchine, gramicidin D, ethidium bromide,
emetine,
mitomycin, procarbazine, mechlorethamine, anthracyclines (e.g., daunorubicin
(formerly
= 5 daunomycin), doxorubicin; doxorubicin liposomal), dihydroxyanthracindione,
mitoxantrone, mithramycin, actinomycin D, procaine, tetracaine, lidocaine,
propranolol,
puromycin, anti-mitotic agents, abrin, ricin A, pseudomonas exotoxin, nerve
growth
factor, platelet derived growth factor, tissue plasminogen activator,
aldesleukin,
allutamine, anastrozle, bicalutamide, biaomycin, busulfan, capecitabine,
carboplain,
chlorabusil, cladribine, cylarabine, daclinomycin, estramusine, floxuridhe,
gamcitabine,
gosereine, idarubicin, itosfamide, lauprolide acetate, levamisole, lomusline,
mechlorethamine, magestrol, acetate, mercaptopurino, mesna, mitolanc,
pegaspergase,
pentoslatin, picamycin, riuxlmab, campath-1, straplozocin, thioguanine,
tretinoin,
vinorelbine, or any fragments, family members, or derivatives thereof,
including
pharmaceutically acceptable salts thereof_ Compositions comprising one or more
chemoagents (e.g., FLAG, CHOP) are also contemplated by the present invention.
FLAG
comprises fludarabine, cytosine arabinoside (Ara-C) and G-CSF. CHOP comprises
cyclophosphamide, vincristine, doxorubicin, and prednisone.
The chemoagent can be an anti-angiogenic agent, such as, for example,
angiostatin,
bevacizumab (Avastin0), sorafenib (Nexavar(D), baculostatin, canstatin,
maspin, anti-
VEGF antibodies or peptides, anti-placental growth factor antibodies or
peptides, anti-Flk-
1 antibodies, anti-Flt-1 antibodies or peptides, laminin peptides, fibronectin
peptides,
plasminogen activator inhibitors, tissue metalloproteinase inhibitors,
interferons,
interleukin 12, IP-10, Gro-(3, thrombospondin, 2-methoxyoestradiol, proliferin-
related
protein, carboxiamidotriazole, CM101, Marimastat, pentosan polysulphate,
angiopoietin 2,
53

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
interferon-alpha, herbimycin A, PNU 145156E, 16K prolactin fragment, Linomide,
thalidomide, pentoxifylline, genistein, TNP-470, endostatin, paclitaxel,
accutin, cidofovir,
vincristine, bleomycin, AGM-1470, platelet factor 4 or minocycline. Without
being bound
by theory, the coadministration of an anti-angiogenic agent advantageously may
lead to
.5 the increase in MN expression in a tumor, thereby making the tumor more
susceptible to
the antibodies and antibody conjugates of the invention.
In one aspect, said chemoagent is gemcitabine at a dose ranging from 100 to
1000
mg/m2/cycle. In one embodiment, said chemoagent is dacarbazine at a dose
ranging from
200 to 4000 mg/ma cycle. In another aspect, said dose ranges from 700 to 1000
mg/m2lcycle. In yet another aspect, said chemoagent is fludarabine at a dose
ranging
from 25 to 50 mg/m2/cycle. In another aspect, said chemoagent is cytosine
arabinoside
(Ara-C) at a dose ranging from 200 to 2000 mg/ m2/cycle. In still another
aspect, said
chemoagent is docetaxel at a dose ranging from 1.5 to 7.5 mg/kg/cycle. In yet
another
aspect, said chemoagent is paclitaxel at a dose ranging from 5 to 15
mg/kg/cycle. In a
further aspect, said chemoagent is cisplatin at a dose ranging from 5 to 20
mg/kg/cycle. In
a still further aspect, said chemoagent is 5-fluorouracil at a dose ranging
from 5 to 20
mg/kg/cycle. In another aspect, said chemoagent is doxorubicin at a dose
ranging from 2
to 8 mg/kg/cycle. In yet a further aspect, said chemoagent is
epipodophyllotoxin at a dose
ranging from 40 to 160 mg/kg/cycle. In yet another aspect, said cheinoagent is
cyclophospharnide at a dose ranging from 50 to 200 mg/kg/cycle. In a further
aspect, said
chemoagent is irinotecan at a dose ranging from 50 to 150 mg/m2/cycle. In a
still further
aspect, said chemoagent is vinblastine at a dose ranging from 3.7 to 18.5
mg/m2/cycle. In
another aspect, said chemoagent is vincristine at a dose ranging from 0.7 to 2
mg/mz/cycle.
In one aspect, said chemoagent is methotrexate at a dose ranging from 3.3 to
1000
mg/m2/cycle.
54

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
In another aspect, the anti-MN antibodies and/or antibody fragments of the
present
invention are administered in combination with one or more immunotherapeutic
agents,
such as antibodies or immunomodulators, which include, but are not limited to,
Herceptin , Retuxan ., OvaRex, Panorex, BEC2, IMC-C225, Vitaxin, Campath I/H,
Smart M195, LymphoCide, Smart -I. D 10, and Oncolym, rituxan, rituximab,
gemtuzumab,
or trastuzumab.
The invention also contemplates administering the anti-MN antibodies and/or
antibody fragments of the present invention with one or more anti-angiogenic
agents,
which includes, but is not limited to, angiostatin, thalidomide, kringle 5,
endostatin, Serpin
(Serine Protease Inhibitor) anti-thrombin, 29 kDa N-terminal and a 40 kDa C-
terminal
proteolytic fragments of fibronectin, 16 kDa proteolytic fragment of
prolactin, 7.8 kDa
proteolytic fragment of platelet factor-4, a(3-amino acid peptide
corresponding to a
fragment of platelet factor-4 (Maione et al., 1990, Cancer Res. 51:2077), a 14-
amino acid
peptide corresponding to a fragment of collagen I (Tolma et al., 1993, J. Cell
Biol.
122:497), a 19 amino acid peptide corresponding to a fragment of
Thrombospondin I
(Tolsma et al., 1993, J. Cell Biol. 122:497), a 20-amino acid peptide
corresponding to a
fragment of SPARC (Sage et al., 1995, J. Cell. Biochem. 57:1329-), or any
fragments,
family members, or derivatives thereof, including pharmaceutically acceptable
salts
thereof.
Other peptides that inhibit angiogenesis and correspond to fragments of
laminin,
fibronectin, procollagen, and EGF have also been described (See the review by
Cao, 1998,
Prog. Mol. Subcell. Biol. 20:161). Monoclonal antibodies and cyclic
pentapeptides, which
block certain integrins that bind RGD proteins (i.e., possess the peptide
motif Arg-Gly-
Asp), have been demonstrated to have anti-vascularization activities (Brooks
et al., 1994,
Science 264:569; Hammes et al., 1996, Nature Medicine 2:529). Moreover,
inhibition of

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
the urokinase plasminogen activator receptor by antagonists inhibits
angiogenesis, tumor
growth and metastasis (Min et al., 1996, Cancer Res. 56:2428-33; Crowley et
al., 1993,
Proc Natl Acad. Sci. USA 90:5021). Use of such anti-angiogenic agents is also
contemplated by the present invention.
. In another=aspect, the anti-MN antibodies and/or antibody fragments of the
present
invention are administered in combination with a regimen of radiation.
The anti-MN antibodies and/or antibody fragments of the present invention can
also be administered in combination with one or more cytokines, which
includes, but is
not limited to, lymphokines, tumor necrosis factors, tumor necrosis factor-
like cytokines,
lymphotoxin-a, lymphotoxin-0, interferon-0, macrophage inflammatory proteins,
granulocyte monocyte colony stimulating factor, interleukins (including, but
not limited
to, interleukin-1, interleukin-2, interleukin-6, interleukin-12, interleukin-
15, interleukin-
18), OX40, CD27, CD30, CD40 or CD137 ligands, Fas-Pas ligand, 4-1BBL,
endothelial
monocyte activating protein or any fragments, family members, or derivatives
thereof,
including pharmaceutically acceptable salts thereof.
The anti-MN antibodies and/or antibody fragments of the present invention can
also be administered in combination with a cancer vaccine, examples of which
include, but
are not limited to, autologous cells or tissues, non-autologous cells or
tissues,
carcinoembryonic antigen, alpha-fetoprotein, human chorionic gonadotropin, BCG
live
vaccine, melanocyte lineage proteins (e.g., gplOO, MART-1/MelanA, TRP-1
(gp75),
tyrosinase, widely shared tumor-associated, including tumor-specific, antigens
(e.g.,
BAGE, GAGE-1, GAGE-2, MAGE-1, MAGE-3, N-acetylglucosaminyltransferase-V,
p15), mutated antigens that are tumor-associated ((3-catenin, MUM-l, CDK4),
nonmelanoma antigens (e.g., HER-2/neu (breast and ovarian carcinoma), human
papillomavirus-E6, E7 (cervical carcinoma), MUC-1 (breast, ovarian and
pancreatic
56

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
carcinoma). For human tumor antigens recognized by T-cells, see generally
Robbins and
Kawakami, 1996, Curr. Opin. Immunol. 8:628. Cancer vaccines may or may not be
purified preparations.
In yet another embodiment, the anti-MN antibodies and/or antibody fragments of
the present invention are used in association with a hormonal -treatment. =
Hormonal
therapeutic treatments comprise hormonal agonists, hormonal antagonists (e.g.,
flutamide,
tamoxifen, leuprolide acetate (LUPRON), LH-RH antagonists), inhibitors of
hormone
biosynthesis and processing, and steroids (e.g., dexamethasone, retinoids,
betamethasone,
cortisol, cortisone, prednisone, dehydrotestosterone, glucocorticoids,
mineralocorticoids,
estrogen, testosterone, progestins), antigestagens (e.g., mifepristone,
onapristone), and
antiandrogens (e.g., cyproterone acetate).
The anti-MN antibodies and/or fragments of the invention can be used in
combination with, e.g. co-administered with, an anti-MDR (multidrug
resistance)
phenotype agent.
Many human cancers intrinsically express or spontaneously develop resistance
to
several classes of anticancer drugs at the same time, notwithstanding that
each of the drug
classes have different structures and mechanisms of action_ This phenomenon,
which can
be mimicked in cultured mammalian cells, is generally referred to as multidrug
resistance
("MDR") or the multidrug resistance phenotype. The MDR phenotype presents
significant
obstacles to the successful chemotherapeutic treatments for cancers in human
patients.
Resistance of malignant tumors to multiple chemotherapeutic agents is a major
cause of
treatment failure (Wittes et al., Cancer Treat. Rep. 70:105 (1986); Bradley,
G. et al.,
Biochim. Biophys. Acta 948:87 (1988); Griswald, D. P. et al., Cancer Treat.
Rep.
65(S2):51 (1981); Osteen, R. T. (ed.), Cancer Manual, (1990)). Tumors
initially sensitive
to cytotoxic agents often recur or become refractory to multiple
chemotherapeutic drugs
57

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
(Riordan et al., Pharmacol. Ther. 28:51 (1985); Gottesman et al., Trends
Pharmacol. Sci.
9:54 (1988); Moscow et al., J. Natl. Cancer Inst. 80:14 (1988); Croop, J. M.
et al., J. Clin.
Invest. 81:1303 (1988)). Cells or tissues obtained from tumors and grown in
the presence
of a selecting cytotoxic drug can result in cross-resistance to other drugs in
that class as
well as other classes. of drugs including, but not-limited to,=
anthracyclines, Vinca alkaloids,
and epipodophyllotoxins (Riordan et al., Pharmacol. Ther. 28:51 (1985);
Gottesman et al.,
J. BioI. Chem. 263:12163 (1988)). Thus, acquired resistance to a single drug
results in
simultaneous resistance to a diverse group of drugs that are structurally and
functionally
unrelated. Such resistance can be a problem for both solid-form and liquid-
form tumors
(e.g. blood or lymph-based cancers).
One major mechanism of multidrug resistance in mammalian cells involves the
increased expression of the 170 kDa plasma membrane glycoprotein pump system
(Juranka et al., FASEB J 3:2583 (1989); Bradley, G. et al., Blochem. Biophys.
Acta
948:87 (1988)). The gene encoding this pump system, sometimes referred to as a
multidrug transporter, has been cloned from cultured human cells and is
generally referred
to as mdrl. This gene is expressed in several classes of normal tissues, but
physiological
substrates transported for the mdri gene product in these tissues have not
been identified.
The MDR1 product is a member of the ABC Transporter Protein superfamily, a
group of
proteins having energy-dependent export function.
The protein product of the mdrl gene, generally known as P-glycoprotein ("P-
170", "P-gp"), is a 170 kDa trans-plasma membrane protein that constitutes the
aforementioned energy-dependent efflux pump. Expression of P-gp on the cell
surface is
sufficient to render cells resistant to multiple cytotoxic drugs, including
many anti-cancer
agents. P-gp-mediated MDR appears to be an important clinical component of
tumor
resistance in tumors of different types, and mdrl gene expression correlates
with
58

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
resistance to chemotherapy in different types of cancer.
The nucleotide sequence of the mdrl gene (Gros, P. et al., Cell 47:371 (1986);
Chen, C. et al., Cell 47:381 (1986)) indicates that it encodes a polypeptide
similar or
identical to P-glycoprotein and that these are members of the highly conserved
class of
membrane proteins siinilar to bacterial transporters -and involved in norrnal
physiological
transport processes. Sequence analysis of the mdrl gene indicates that Pgp
consists of
1280 amino acids distributed between two homologous (43% identity) halves.
Each half
of the molecule has six hydrophobic transmembrane domains and each has an ATP
binding site within the large cytoplasmic loops. Only about 8% of the molecule
is
extracellular, and the carbohydrate moiety (approximately 30 kDa) is bound to
sites in this
region.
Thus, it will be appreciated that mammalian cells having a "multidrug-
resistance"
or "multidrug-resistant" phenotype are characterized by the ability to
sequester, export or
expel a plurality of cytotoxic substances (e.g., chemotherapeutic drugs) from
the
intracellular milieu. Cells may acquire this phenotype as a result of
selection pressure
imposed by exposure to a single chemotherapeutic drug (the selection toxin).
Alternatively, cells may exhibit the phenotype prior to toxin exposure, since
the export of
cytotoxic substances may involve a mechanism in connnon with normal export of
cellular
secretion products, metabolites, and the like. Multidrug resistance differs
from simple
acquired resistance to the selection toxin in that the cell acquires
competence to export
additional cytotoxins (other chemotherapeutic drugs) to which the cell was not
previously
exposed. For example, Mirski et al. (1987), 47 Cancer Res. 2594-2598, describe
the
isolation of a multidrug-resistant cell population by culturing the H69 cell
line, derived
from a human small cell lung carcinoma, in the presence of adriamycin
(doxorubicin) as a
selection toxin. Surviving cells were found to resist the cytotoxic effects of
anthracycline
59

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
analogs (e.g., daunomycin, epirubicin, menogaril and mitoxantrone), acivicin,
etoposide,
gramicidin D, colchicine and Vinca-derived alkaloids (vincristine and
vinblastine) as well
as of adriamycin. Similar selection culturing techniques can be applied to
generate
additional multidrug-resistant cell populations.
= 5 Accordingly, the pharmaceutical compositions of the invention can
additionally
include compounds which act to inhibit the MDR phenotype and/or conditions
associated
with MDR phenotype. Such compounds can include any known MDR inhibitor
compounds in the art, such as, antibodies specific for MDR components (e.g.
anti-MDR
transporter antibodies) or small molecule inhibitors of MDR transporters,
including
specifically, tamoxifen, verapamil and cyclosporin A, which are agents known
to reverse
or inhibit multidrug resistance. (Lavie et al. J. Biol. Chem. 271: 19530-
10536, 1996,
incorporated herein by reference). Such compounds can be found in U.S. Patents
Nos.
5,773,280, 6,225,325, and 5,403,574, each of which are incorporated herein by
reference.
Such MDR inhibitor compounds can be co-administered with the anti-MN
antibodies
and/or fragments of the invention for various purposes, including, reversing
the MDR
phenotype following the detection of the MDR phenotype to assist or enhance a
chemotherapeutic treatment. The MDR inhibitor, such as, for example,
tamoxifen,
verapamil or cyclosporin A, may be used, in conjuction with the compounds of
the
invention to assist in the detection of the MDR phenotype. In accordance with
this aspect,
an MDR inhibitor can enhance the uptake and accumulation of a compound of the
invention in an MDR cancer cell since the capacity of the MDR transport system
in
transporting or "pumping out" the imaging compound vis-a-vis the substrate
domain
would be diminished in the presence of an MDR inhibitor.
In yet another embodiment, the anti-MN antibodies and/or antibody fragments of
the present invention are used in association with a gene therapy program in
the treatment

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
of cancer. Gene therapy with recombinant cells secreting interleukin-2 can be
administered in combination with the inventive antibodies to prevent or treat
cancer,
particularly breast cancer (See, e.g., Deshmukh et al., 2001, J. Neurosurg.
94:287).
To assess the ability of a particular antibody to be therapeutically useful to
treat
cancer, as an example, the antibody may be tested in vivo in a mouse xenograft
tumor
model. If desired, MN antibodies may be converted into IgG, antibodies before
therapeutic assessment. This conversion is described in Example 5, and an
example of a
therapeutic model is detailed in Example 9. Antibody activity may also be
tested using an
antibody dependent cell-mediated cytotoxicity assay as described in Example
12.
The present invention also provides diagnostic methods with which MN may be
detected in a patient sample or biological sample. Such diagnostic methods may
be used,
for example, to diagnose disorders in which MN is elevated. Such disorders
include, but
are not limited to, carcinomas of the kidney, esophagus, breast, cervix,
colon, and lung.
When used for diagnosis, detection of an amount of the antibody-MN complex in
a sample
from a patient which is greater than an amount of the complex in a normal
sample
identifies the patient as likely to have the disorder. An immunohistochemical
method for
the detection of MN in cancer tissues is described in Example 11.
In another aspect of the invention, a method is provided for detecting and/or
visualizing an MN-related cancer having an abnormally amount of expressed MN
protein.
Such methods can comprise contacting the MN-related cancer cell with an anti-
MN
antibody or fragment of the invention, and making an image using a medical
imaging
modality, wherein the anti-MN antibody or fragment comprises a label domain
capable of
being detected by the medical imaging modality.
The detection methods of the invention can be performed in vitro. The cancer
cell
or tissue can be from any suitable source, such as for example, a biopsy or a
cell or tissue
61

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
culture. Methods for obtaining biopsies and maintaining and/or propagating the
removed
tissues and/or cells will be well known to the skilled artisan. In vitro
detection of
multidrug resistance can have various applications, such as, for example,
determining
whether a particular subject's cancer, either before, during or after
treatment, has
== 5 developed a multidrug phenotype. -
The type of imaging modality used to detect the compounds of the invention
will
depend on the particular label domain used in the inventive compounds. For
example, if
the label domain comprises a gadolinium chelate, then typically MRI could be
used to
detect the imaging agent of the invention. If a radionuclide chelate is used
as the label
domain, a nuclear imaging method could be used (e.g. PET). If a fluorescence-
based label
domain is used, an optical imaging system could be used, such as, for example
a FACS
system or fluorescence microscopy or a fluorescence automated plate reader.
Choosing an
appropriate imaging modality for use in the in vitro detection methods of the
invention are
completely within the knowledge of the skilled artisan.
In addition, the amount of imaging agent used in the in vitro detection
methods of
the invention will be determined by one of ordinary skill in the art and can
depend on the
degree to which the MDR phenotype is present, e.g. the level of expression of
the MDR
transport system (e.g. the P-glycoprotein). The skilled artisan can determine
what amount
of the novel imaging compounds that is sufficient for detecting a MDR
phenotype without
undue experimentation, i.e. a detectably sufficient amount.
The type of imaging modality used is not limited to any particular type, and
can
include, for example MRI, nuclear imaging (e.g. PET or SPECT), optical
imaging,
sonoluminence imaging or photoacoustic imaging (ultrasound). The skilled
artisan will
appreciate that the particular label domain of the imaging compounds of the
invention
should be compatible with the particular imaging modality being used.
62

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
In a preferred embodiment, the methods of detection utilize anti-MN antibodies
or
fragments thereof and appropriate labels that are capable of being detected by
MRI. For
example, the antibodies or fragments of the invention can comprise a label
domain that is
a MR contrast agent, such as, for example a paramagnetic metal chelate or
chelates or any
.5 of those described herein: The imaging agent can also comprise a
radionuclide label
domain for imaging or detecting by a nuclear imaging modality, such as,
positron emission
tomography (PET) or single photon emission computer tomography (SPECT), e.g. a
radionuclide such as, for example, 199Au, 72 As, 141 Ce, 67Cu, 60Cu, 5aFe,
67Ga, 68Ga, S~Gr,
I I 1In, I77Lu, SIMn, 203Pb, 18sRe, 9'7Ru, 47SC, 177msn, 94m.rC, 167Tm, and
90Y. The
radionuclide can be chelated by a suitable chelator, or by multiple chelators,
such as, for
example HYNIC, DRPA, EDTA, DOTA, TETA, DTPA and BAT. Conditions under
which a chelator will coordinate a metal are described, for example, by Gansow
et al., U.S.
Pat. Nos. 4,831,175, 4,454,106 and 4,472,509, each of which are incorporated
herein by
reference. 99mTc (Technetium-99m) is a particularly attractive radioisotope
for therapeutic
and diagnostic applications, as it is generally available to nuclear medicine
departments, is
inexpensive, gives minimal patient radiation doses, and has ideal nuclear
imaging
properties.
The patient sample may be contacted with an antibody of the invention, and the
patient sample may then be assayed for the presence of an antibody-MN complex.
As
described above, the antibody may comprise a detectable label, such as a
fluorescent,
radioisotopic, chemiluminescent, or enzymatic label, such as horseradish
peroxidase,
alkaline phosphatase, or luciferase.
Optionally, the antibody may be bound to a solid support, which may
accommodate automation of the assay. Suitable solid supports include, but are
not limited
to, glass or plastic slides, tissue culture plates, microtiter wells, tubes,
silicon chips, or
63

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
particles such as beads (including, but not limited to, latex, polystyrene, or
glass beads).
Any method known in the art may be used to attach the antibody to the solid
support,
including use of covalent and non-covalent linkages, passive absorption, or
pairs of
binding moieties attached to the antibody and the solid support. Binding of MN
and the
antibody may be accomplished in= any vessel suitable for containing the
reactants.
Examples of such vessels include microtiter plates, test tubes, and
microcentrifuge tubes.
Pharmaceutical Compositions and Dosages
The antibodies described herein may be provided in a pharmaceutical
composition
comprising a pharmaceutically acceptable carrier. The pharmaceutically
acceptable
carrier may be non-pyrogenic. The compositions may be administered alone or in
combination with at least one other agent, such as stabilizing compound, which
may be
administered in any sterile, biocompatible pharmaceutical carrier including,
but not
limited to, saline, buffered saline, dextrose, and water. A variety of aqueous
carriers may
be employed including, but not limited to saline, glycine, or the like. These
solutions are
sterile and generally free of particulate matter. These solutions may be
sterilized by
conventional, well-known sterilization techniques (e.g., filtration).
Generally, the phrase "pharmaceutically acceptable carrier" is art recognized
and
includes a pharmaceutically acceptable material, composition or vehicle,
suitable for
administering compounds of the present invention to ma.mmals. The carriers
include
liquid or solid filler, diluent, excipient, solvent or encapsulating material,
involved in
carrying or transporting the subject agent from one organ, or portion of the
body, to
another organ, or portion of the body. Each carrier must be "acceptable" in
the sense of
being compatible with the other ingredients of the formulation and not
injurious to the
patient. Some examples of materials which can serve as pharmaceutically
acceptable
carriers include: sugars, such as lactose, glucose and sucrose; starches, such
as corn starch
64

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
and potato starch; cellulose, and its derivatives, such as sodium
carboxymethyl cellulose,
ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin;
talc; excipients,
such as cocoa butter and suppository waxes; oils, such as peanut oil,
cottonseed oil,
safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such
as propylene
glycol; polyols, such as glycerin; sorbitol, mannitol and polyethylene glycol;
esters, such
as ethyl oleate and ethyl laurate; agar; buffering agents, such as magriesium
hydroxide and
aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline;
Ringer's solution;
ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible
substances
employed in pharmaceutical formulations.
Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and
magnesium stearate, as well as coloring agents, release agents, coating
agents, sweetening,
flavoring and perfuming agents, preservatives and antioxidants can also be
present in the
antibody compositions of the invention.
Examples of pharmaceutically acceptable antioxidants include: water soluble
antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate,
sodium
metabisulfite, sodium sulfite and the like; oil-soluble antioxidants, such as
ascorbyl
palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT),
lecithin,
propyl gallate, alpha-tocopherol, and the like; and metal chelating agents,
such as citric
acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid,
phosphoric acid, and
the like.
The compositions may contain pharmaceutically acceptable auxiliary substances
as
required to approximate physiological conditions such as pH adjusting and
buffering
agents, and the like. The concentration of the antibody of the invention in
such
pharmaceutical formulation may vary widely, and may be selected primarily
based on
fluid volumes, viscosities, etc., according to the particular mode of
administration

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
selected. If desired, more than one type of antibody may be included in a
pharmaceutical
composition (e.g., an antibody with different Kd for MN binding).
The compositions may be administered to a patient alone, or in combination
with
other agents, drugs or hormones. In addition to the active ingredients, these
.5 pharmaceutical compositions may contain suitable pharmaceutically
acceptable carriers
comprising excipients and auxiliaries that facilitate processing of the active
compounds
into preparations which may be used pharmaceutically. Pharmaceutical
compositions of
the invention may be administered by any number of routes including, but not
limited to,
oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal,
intraventricular,
transdermal, subcutaneous, intraperitoneal, intranasal, parenteral, topical,
sublingual, or
rectal means.
The compositions of the invention additionally contemplate suitable
immunocarriers, such as, proteins, polypeptides or peptides such as albumin,
hemocyanin,
thyroglobulin and derivatives thereof, particularly bovine serum albumin (BSA)
and
keyhole limpet hemocyanin (KLH), polysaccharides, carbohydrates, polymers, and
solid
phases. Other protein-derived or non-protein derived substances are known to
those
skilled in the art.
In aspects involving vaccines, e.g. cancer vaccines together with the
antibodies of
the invention, the compositions of the invention can be administered with or
without an
adjuvant. Administration can be carried out in the absence of an adjuvant in
order to avoid
any adjuvant-induced toxicity. The person of ordinary skill in the art to
which this
invention pertains, e.g. a medical doctor specializing in cancer, will
appreciate and
understand how to ascertain whether an adjuvant should or should not be used
and can
dependent upon the medical history of a subject, family data, toxicity data,
allergy-related
test results, etc. In embodiments where an adjuvant is used, it is
advantageous that the
66

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
adjuvant promotes the formation of protective antibodies, such as protective
IgG
antibodies. Any suitable adjuvant known to one of ordinary skill in the art is
contemplated
by the present invention and are readily adapted to this invention. Suitable
adjuvants for
use in vaccinating animals can include, but are not limited to, aluminum
hydroxide,
.==5 aluminum hydroxide, saponin-and-its purified component Quit A, complete
Freund's
adjuvant (CFA) and incomplete Freund's adjuvant (IFA). Dextran sulfate has
been shown
to be a potent stimulator of IgG2 antibody against staphylococcal cell surface
antigens, and
also is suitable as an adjuvant. It will be appreciated by the skilled person
that some
adjuvants can be more preferable for veterinary application, whereas other
adjuvants will
be preferable for use in humans, and that adjuvant toxicities are a
consideration that should
be made by the skilled person prior to administration of the compound to a
human.
Formulations suitable for parenteral, subcutaneous, intravenous,
intramuscular, and
the like; suitable pharmaceutical carriers; and techniques for formulation and
administration may be prepared by any of the methods well known in the art
(see, e.g.,
Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 20'h
edition,
2000). Liquid dosage forms for oral administration of the compounds of the
invention
include pharmaceutically acceptable emulsions, microemulsions, solutions,
suspensions,
syrups and elixirs. In addition to the active ingredient, the liquid dosage
forms may
contain inert diluent commonly used in the art, such as, for example, water or
other
solvents, solubilizing agents and emulsifiers, such as ethyl alcohol,
isopropyl alcohol,
ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene
glycol, 1,3-
butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ,
olive, castor and
sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and
fatty acid esters
of sorbitan, and mixtures thereof.
67

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
The phrases "parenteral administration" and "administered parenterally" as
used
herein means modes of administration other than enteral and topical
administration,
usually by injection, and includes, without limitation, intravenous,
intramuscular,
intraarterial, intrathecal, intracapsular, intraorbital, intracardiac,
intradermal,
=5 intraperitoneal, transtracheal, subeutaneous, subcuticular, intraarticular,
subcapsular,
subarachnoid, intraspinal and intrasternal injection and infusion.
The determination of a therapeutically effective dose is well within the
capability
of those skilled in the art. A therapeutically effective dose refers to the
amount of an
antibody that may be used to effectively treat a disease (e.g., cancer)
compared with the
efficacy that is evident in the absence of the therapeutically effective dose.
The therapeutically effective dose may be estimated initially in animal models
(e.g., rats, mice, rabbits, dogs, or pigs). The animal model may also be used
to determine
the appropriate concentration range and route of administration. Such
information may
then be used to determine useful doses and routes for administration in
humans.
Therapeutic efficacy and toxicity (e.g., ED50 - the dose therapeutically
effective in
50% of the population and LD50 - the dose lethal to 50% of the population) of
an antibody
may be determined by standard pharmaceutical procedures in cell cultures or
experimental
animals. The dose ratio of toxic to therapeutic effects is the therapeutic
index, and it may
be expressed as the ratio, LD50/ED50. The data obtained from animal studies
may used in
forrnulating a range of dosage for human use. The dosage contained in such
compositions
may be within a range of circulating concentrations that include the ED50 with
little or no
toxicity. The dosage varies within this range depending upon the dosage form
employed,
sensitivity of the patient, and the route of administration.
The exact dosage may be determined by the practitioner, in light of factors
related
to the patient who requires treatment. Dosage and administration may be
adjusted to
68

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
provide sufficient levels of the antibody or to maintain the desired effect.
Factors that rnay
be taken into account include the severity of the disease state, general
health of the subject,
age, weight, and gender of the subject, diet, time and frequency of
administration, drug
combination(s), reaction sensitivities, and tolerance/response to therapy.
Polynucleotides encoding antibodies of-the invention may be constructed and
introduced into a cell either ex vivo or in vivo using well-established
techniques including,
but not limited to, transferrin-polycation-mediated DNA transfer, transfection
with naked
or encapsulated nucleic acids, liposome-mediated cellular fusion,
intracellular
transportation of DNA-coated latex beads, protoplast fusion, viral infection,
electroporation, "gene gun," and DEAE- or calcium phosphate-mediated
transfection.
Effective in vivo dosages of an antibody are in the range of about 5 g to
about 500
g/kg of patient body weight. For administration of polynucleotides encoding
the
antibodies, effective in vivo dosages are in the range of about 100 ng to
about 500 .g of
DNA.
The mode of administration of antibody-containing pharmaceutical compositions
of the present invention may be any suitable route which delivers the antibody
to the host.
As an example, pharmaceutical compositions of the invention may be useful for
parenteral
administration (e.g., subcutaneous, intramuscular, intravenous, or intranasal
administration).
All patents and patent applications cited in this disclosure are expressly
incorporated herein by reference_ The above disclosure generally describes the
present
invention. A more complete understanding can be obtained by reference to the
following
specific examples, which are provided for purposes of illustration only and
are not
intended to limit the scope of the invention.
EXAMPLES
69

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
The structures, materials, compositions, and methods described herein are
intended
to be representative examples of the invention, and it will be understood that
the scope of
the invention is not limited by the scope of the examples. Those skilled in
the art will
recognize that the invention may be practiced with variations on the disclosed
structures,
= 5 materials, compositions and methods, and such variations are regarded as
within the ambit
of the invention
Example 1: Construct of Human Combinatorial Fab library (HuCAL Gold)
HuCAL GOLD is an antibody library based on HuCAL technology (Human
Combinatorial Antibody Library; MorphoSys AG, Martinsried/Planegg, Germany).
The
library combines a synthetic, fully human antibody library in the Fab format
featuring six
diversified CDR regions with an display technology for the selection of high
affinity
antibodies, CysDisplayTM (MorphoSys AG, Martinsried/Planegg, Germany).
CysDisplayTM is a monovalent phage display technology based on a phenotype-
genotype
linkage via disulfide bonds, which allows the recovery of specific antibodies
with high
affinities.
The phage display vector pMORPH 23 is a phagemid vector allowing monovalent
CysDisplayTM of Fab fragments. It encodes the full length gIIIp, the Fd chain
(VH-CH1),
and the light chain (VL-CL), all of which are equipped with different
secretory signal
sequences which directs the corresponding protein chain to the periplasm of E.
coli. Both
ompA and phoA signal sequences are utilized to transport the heavy and light
chains to the
periplasm, where the chains assemble via non-covalent interactions occurs.
This display
vector carries an inducible lac promotor/operator region. The laqlq gene
product for
repression of expression is supplied by the E. coli host strain TG1. Induction
of Cys-gIIIp
and Fab expression is achieved by the addition of IPTG (isopropyl-J3-D-thio-
galacto-

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
pyranoside). The enriched Fab pools were sub-cloned from pMORPH 23 to the Fab
expression vector pMORPH x9 using the restriction enzymes Xbal and EcoRl. By
this
step, the cysteine and the linker-His6 part at the C-terminus of the Fd chain
were removed
and the gIIIp is excised. The expression vector pMORPH x9_Fab_FH provides two
C-
terminal tags a FLAG -and.6xHis thus facilitating=detection and purification
of the Fab
proteins.
Phagemid rescue and phage amplication
The HuCAL GOLD Fabs phagemid in TG1 cells were amplified in 2xTY media
supplemented with 34 g/ml chloramphenicol and 1% glucose. After helper phage
infection (VCSM13 -r2-6x 10" pfu/ml) at 37 C for 30 minutes, TGl cells were
concentrated by centrifugation_ The phage were amplified by incubation of the
infected
TG1 cells at 22 C overnight in 2xTY media containing 34 p,g/ml
chloramphenicol, 50
g/rni kanamycin, and 0.25 mM IPTG. The phage containing supernatant was used
for
the phase panning.
Example 2: Solid phase panning
Solid phase panning was carried out by coating MaxiSorpTM plates (Nalgene Nunc
International, Rochester, NY) or DynabeadsTM (Invitrogen, Carlsbad, CA) with
human
MN protein in PBS (1 g/well or 1 g/1 mg of beads). The MN protein represents
the
entire extra-cellular domain of the protein with a C-terminal histag for
purification. The
MN protein was expressed in a mammalian cell line HKB-11 and purified by Ni-
NTA
chromatography using standard methods that are well known to those skilled in
the art.
Wells containing bound MN protein were blocked with 5% mi.lk in PBS, washed in
PBS
followed by a 2 hour incubation at room temperature with an aliquot of pre-
blocked
HuCAL GOLD phage library containing lxl01a HuCAL GOLD Fab phage. Bound phage
were washed and then eluted with 20 mM DTT in 10 mM Tris buffer at pH 8Ø
Three
71

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
rounds of panning were performed with phage amplification conducted between
each
round as described above. Wash stringency was increased between each round of
panning
to decrease nonspecific binding.
Example 3: Subcloning of selected Fab fragments for expression in E. coli
.= 5 The selected Fabs were cloned from the pMORPHO23=display vector to the
pMORPHOx9 Fab_FH expression vector to facilitate rapid expression of soluble
Fab for
ELISA screening. The DNA preparation of the pMORPH 23 vector was digested with
EcoRI and Xbal, thus cutting out the entire Fab-encoding fragment (ompA-VL-VL
and
phoA-Fd). After subsequent purification, the fragment was ligated to the
prepared
pMORPHx9_Fab_FH vector (also digested with EcoRI/XbaI and purified). The
vector
containing the Fab insert was transfected into competent E.coli TG1 F- cells
by
electroporation. The transformed E. coli cell were grown on LB plates
containing 34
gg/m1 chloramphenicol and i% glucose o/n at 37 C. Colonies were picked and
placed in
culture media containing 34 g/ml chloramphenicol and 1% glucose. Glycerol was
added
to 20% and these stock starter cultures were stored at -80 C until needed for
ELISA. To
obtain purified Fabs, E.coli transformants carrying this vector were grown
typically in
multiples of 1L shake flask cultures, harvested, and purified using Ni-NTA
affinity
chromatography using methods well known to those skilled in the art.
Example 4: Identification of MN-Binding Fab fragments by ELISA
Maxisorp 96-well ELISA plates were coated with 100 l/well of a solution of
purified human MN protein at a concentration of 5 .g/nil in PBS. Fabs were
expressed
using E.coli transformants grown in 2xTY media containing 34 g/ml
chloramphenicol
from started stock cultures. Twelve (12) hours prior to harvest Fab expression
was
induced by addition of 1PTG (0.5 mM final concentration). Cells were lysed and
100 l
lysate was incubated within the well of an MN-precoated and milk-blocked
Maxisorp
72

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
plate for 2 hours at room temperature. Plates were then washed in TBS and
Tween
containing-TBS to remove nonspecific binding. Bound Fabs were detected with
goat anti-
human (Fab')2 antibody conjugated to alkaline phosphatase (Pierce Chemical,
Rockford,
IL). The substrate AttoPhos (Roche Diagnostics, Alameda, CA) was used as
directed in
the manufacturers instructions, excitation was at 430 nm and emission was read
at 535 nm.
A large number of E.coli transformants expressed Fabs that exhibited a signal
to
noise ration in the ELISA of > 10. DNA sequencing of the VH and VL regions
identified
over 50 unique MN-binding Fabs. The DNA and protein sequences of the entire VH
and
VL of ten antibodies based upon their functional properties is shown in
Figures 3 and 4,
respectively. Each of these ten disclosed antibodies bound by ELISA to the
purified MN
protein isolated from a mamrnalian expression cell line, HKB-1 1 (Figure 5).
The
antibodies examined also reacted with purified MN isolated from sf9 insect
cells that had
been infected with a baculovirus encoding the extra-cellular domain of MN,
(Figure 5)
demonstrating that the ELISA interactions were specific for the MN protein and
not any
trace contaminants in the protein preparations. In a BIAcoreTM assay,
antibodies of the
present invention specifically bind to human MN with a Kd in the range from 1
nM (1 x
10'9 M) to about 50 nM (5.0 x 10-$ nM) (Figure 5).
Example 5: Identification of MN-binding Fabs and IgGs
The binding interactions between MN protein and the antibodies of this
invention
were analyzed using surface plasmon resonance technology on a BlAcore 3000
instrument
(BIAcore, Uppsala, Sweden). For the binding of full-length IgGs (1e4, laal,
and 3ee9),
goat anti-human IgG Fc was covalently coupled to the CM5 biosensor chip at
high density
using a standard amine coupling kit (BlAcore, Uppsala, Sweden). A capture
assay was
then used to bind these IgG to the anti-human IgG Fc-immobilized surface,
aiming for a
300 response unit BlAcore signal. The BlAcore was operated at 25 C and a flow
rate of
73

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
1/min running buffer containing PBS/0.05% Tween 20, or containing 10mM Na-
HEPES, pH 7.5/150mM NaCU3mM EDTA/0.005% Tween 20. After ligand binding, the
flow rate was then increased to 25 Umin and various concentrations of MN
analyte (e.g.,
range of 50 nM to 1 nM) were flowed over the surface for 10 minutes such that
the
5 association phase could be monitored. Dissociation then proceeded for 35
minutes,
followed by regeneration of the anti-human IgG Fc-immobilized surface at 100
l/min
with 10011.1 of 10 mM phosphoric acid. Sensorgrams were fitted using a 1:1
Langmuir
binding model to calculate rate constants. The binding of Fabs to MN was
similarly
determined except that an anti-human-Fab IgG was immobilized onto the chip and
used to
10 capture the Fabs prior to the assessment of MN binding. Figure 5 shows the
resulting
binding constants for the binding of the disclosed antibodies to purified MN
protein. Each
of the antibodies bound MN exhibiting Kd values that ranged between 0.15 and
50 nM.
Example 6: Construction of HuCAL immunoglobulin expression vectors for
transient expression antibody expression in 293 F cells
Heavy chain expression vector. The multiple cloning site of pcDNA3.1+
(Invitrogen, Carlsbad, CA) was removed (NheI/Apal), and a site compatible with
the
restriction sites used for HuCAL was inserted for the ligation of the leader
sequences
(Nhei/EcoRI), VH-domains (EcoRI/B1pI), and the immunoglobulin constant regions
(Blpl/ApaI). The leader sequence (EMBL M83133) was equipped with a Kozak
sequence
(Kozak, Nucleic Acid Res. 15:8125-8148,1987). The constant regions of human
IgGl
(PIR J00228), IgG4 (EMBL K01316), and serum IgA 1(EMBL J00220) were dissected
into overlapping oligonucleotides with lengths of about 70 bases. Silent
mutations were
introduced to remove restriction sites non-compatible with the HuCAL design.
The
oligonucleotides were spliced by overlap extension-PCR.
74

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Light chain expression vectors. The multiple cloning site of pcDNA3.1/Zeo+
(Invitrogen, Carlsbad, CA) was replaced by two different sites. The K-site
provided
restriction sites for insertion of a ic-leader (NheI/EcoRV), HuCAL-scFv Vk-
domains
(EcoRVBsiWI,) and the K-chain constant region (BsiWI/ApaI). The corresponding
restriction sites in the a,-site were Nhel/EcoRV (1-leader), EcoRV/HpaI (Vl-
domains), and
HpaI/Apal (X-chain constant region). The x-leader (EMBL Z00022) as well as the
;~-
leader (EMBL L27692) were both equipped with Kozak sequences. The constant
regions
of the human x-chain (EMBL J00241) and X-chain (EMBL M18645) were assembled by
overlap extension-PCR as described above.
Generation offull-length IgG from Fabs. Fab heavy chain sequence contained
within the E. coli expression vector pMORPHx9_Fab_FH was excised by cutting
with
MfeIIB1pI and ligated into the heavy chain expression vector described above
that had
been cut with EcoRI/B1pI. Fab kappa light chain sequence contained within
pMORPHx9_Fab_FH was excised with EcoRV/BsiWI and ligated into the Kappa light
chain expression vector described above that had also been cut with
EcoRV/BsiWI. Fab
lambda light chain sequence contained within pMORPHx9_Fab_FH was excised with
EcoRV/Hpal and ligated into the lambda expression vector described above.
Large scale transient expression offull-length IgGs. Ceilbag 20L/0 (Wave
Biotech LLC, Somerset, NJ) were seeded with 0.25 x 106 293F cells/ml
(Invitrogen) in 9.3
L Freestyle 293 expression medium (Invitrogen). Cells were grown to a density
of 1
x 106/ml and transfected by the addition of 5 mg each of the light chain
expression vector
and heavy chain expression vector encoding the full length antibody in 350 ml
Optimern
(Invitrogen) containing 293fectin reagent (Invitrogen). Following fermentation
for 96
hours at 37 C, cell culture supernatant was harvested by centrifugation,
sterile filtered,

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
concentrated by tangential flow filtration adjusted to pH 7.6 and then
subjected to standard
protein A column chromatography (Amersham Pharmacia Biotech, Piscataway, NJ).
Example 7: Cell Adhesion Assay
Fifty (50) I.t.L of a 1 gg/mL solution of purified MN in PBS was adsorbed onto
a
non-treated 96-well plate overnight at 4 C. The solution was removed,-and the
wells
rinsed 3x with PBS. The wells were blocked for 1 hour with 200 pL 50% FBS in
RPMI1640 media. The wells were then treated with 100 g 1e4 anti-MN antibody
in 1%
BSA in PBS, control IgG in 1% BSA in PBS or 1% BSA in PBS. After washing with
PBS, 5000 MaTu cells (MN+ cells) were added to the wells and the plate
incubated
overnight at 37 C at 5% CQ2. The ability of anti-MN antibodies to block
adhesion of
MaTu cells to MN-coated wells was assessed after washing with PBS. An example
of this
experiment is shown in Figure 6 where 100 g of anti-MN antibody 1e4 inhibits
cell
adhesion, whereas control IgG or the buffer vehicle does not.
Example 8: Subcutaneous Xenograft Cancer Model with Immunoconjugate
Anti-MN antibodies were conjugated to cytotoxic small molecules using
protocols
that are known in the art (e.g., Liu, et al., Proc. Natl. Acad. Sci. 93:8618-
8623, 1996.).
Human mammary xenograft, MaTu cells were maintained as adherent cultures in
RPMI
supplemented with 10% FBS. Ncr nude mice (8-12 weeks of age) were inoculated
subcutaneously in the right flank with 5 x 106 cells in 0.1 mL of 80%
matrigel/20% HBSS.
When tumors reached an average size of -180 mg (6 days), treatment was
initiated.
Monoclonal antibodies conjugated to cytotoxic small molecules were
administered i.v.
once every four days (Q4Dx3) at a dose of 10 mg/kg. Control mice were treated
with PBS
or an unconjugated monoclonal antibody. Daily examinations into the health
status of
each animal were conducted. Each experimental group consisted of 10 mice and
the
dosing volume was 0.1 mL/10 g body weight. The length and width of each tumor
was
76

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
measured by using an electronic caliper 2-3 times per week and tumor weights
(mg) were
calculated based on the formula of [length (mm) x width (mm)2]/2. All data,
including
daily observations, obtained throughout the course of the study were
documented. Tumor
growth inhibition (TGI) was calculated as 1-T/Cx100, where T = final tumor
weights from
a treated group, and C-.final tumor weights from the control group. Figure 7
shows that
the monoclonal IgGi 1e4 when conjugated to a cytotoxic drug produced a
significant anti-
tumor effect at 30 mg/kg imrnunoconjugate, whereas the unconjugated antibody
had no
effect. These data demonstrate that the therapeutic utility of antibodies
directed against
MN protein as vehicles for cytotoxic drug delivery to tumors.
Example 9: Fluorescence-activated cell sorting assay (FACS assay)
Cells may be assayed for MN expression as a diagnostic tool. Adherent MN-
expressing PC-3 mm2 cells and non-MN expressing DLD1 cells were detached from
their
flasks with 1:10 trypsin/Versene in PBS solution for 5 to 10 minutes. Cells
were spun
down (1000 rpm, 5 minutes), washed once with ice cold RPMI 10% FBS, and
resuspended
in ice-cold staining buffer(Ca+ Me-free PBS, 2% BSA, and 0.05% sodium azide)
at 6 x
106 cells/ml. Primary antibody, human anti-MN IgG1, or control human IgG 1
antibody at
g/mL were incubated with 6 x 105 cells on ice for 1 hour. The unbound antibody
was
washed from the cells with the ice-cold staining buffer. The cells were fixed
with 2%
formaldehyde in PBS for 10 minutes, then washed twice with staining buffer.
The cell
20 pellet was resuspended in 100 p.1 ice cold staining buffer containing anti-
human Alexa
fluor 488 secondary antibody (final conc. 1:200, Molecular Probes/Invitrogen,
Carlsbad,
CA), and incubated on ice for 1 hour. The unbound antibody was washed from the
cells
two times with flow buffer (PBS containing 2% BSA), and the cells were
resuspended in 1
mL flow buffer. FACS analysis of the resuspended cells was performed on a
Beckman
77

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
FACS Caliber instrument. Figure 8 shows that PC-3 mm2 human prostate cancer
cells
expressed MN as assayed by FACS, whereas DLD-1 cells did not.
Example 10: Antibody-dependent cell mediated cytotoxicity assays (ADCC assays)
Anti-tumor activity of anti-MN IgGs may be mediated by ADCC activity. MN-
expressing PC-3 mm2 cells and non-MN expressing HCT-1 16 cells are-incubated
with
250 ng/mL, 1000 ng/mL, or 2000 ng/mL human anti-MN IgG 1, or control human IgG
1
anti-digoxin antibody. Human PBMCs are added to these cells at effector:
target ratios of
50:1, 25:1, and 5:1 ratios. A chromium-51 release assay is performed to
determine the
level of target cell lysis. A small amount of lysis is observed upon
incubation of control
antibody or no antibody in the presence of DLD and PC-3 mm2 cells. This
spontaneous
level of lysis is 10-15%, 5-10%, or 2-3% for 50:1, 25:1, and 5:1 target
effector ratios,
respectively. Similarly, lysis of non-MN expressing DLD cells was in the 0-10%
range
when incubated with the anti-MN antibodies. However, lysis of PC-3 mm2 cells
when
incubated with the human anti-MN IgGs was significantly higher than the
controls. Lysis
of 40, 50, and 60% was observed when using 250 ng/mL, 1000 ng/mL, and 2000
ng/niL at
50:1 target:effector ratios. Similarly, 30, 33, and 38% lysis was observed at
25:1 ratios,
and finally, 8, 10, and 15% lysis was observed at 5:1 target:effector ratios.
These
experiments show that human anti-MN antibodies mediate anti-tumor ADCC
activity and
may be used for the therapeutic treatment of cancer.
Example 11. Immunoconiugate
Preparation
A human antibody directed at the MN cell surface antigen was generated using a
phage display library that encodes a diversity of human Fabs (Morphosys). This
antibody
was conjugated to Monomethylauristatin E (MMAE) (Fransisco et al., Blood,
2003,
102:1458-1465) (Figure 9).
78

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
In Vitro Activity and Selectivity
The antibody portion of 3ee9/MMAE (3ee9) was identified by in vitro "panning"
of a purified extracellular domain of human MN against the MorphoSys phage
library
composed of 1010 human Fab fragments (Fabs are the antigen binding portions of
-5 antibodies). The active Fabs were =further examined for their capacity to
=selectively bind
and undergo internalization upon addition to MN positive cells. The resulting
active Fabs
were then converted to full length human IgGl antibodies, expressed in CHO
cells,
purified and then conjugated to the toxophore, MMAE (Liu et al, Proc Natl Acad
Sci,
1996, 93:8618-8623). The conjugated antibodies were then tested for their
ability to kill
MN expressing cells. From a panel of seven full-length antibodies tested,
3ee9/MMAE
was selected based on its binding properties, selectivity and potency in both
in vitro and in
vivo assays.
Surface Plasmon Resonance (Biacore) Materials and Methods
Binding interactions between HKB1 1-expressed human MN protein and human
full-length anti-MN MAbs were analyzed using surface plasmon resonance
technology on
a BlAcore 3000 instrument (BIAcore, Uppsala, Sweden). For chip preparation,
goat anti-
human IgG Fc was covalently coupled to the CM5 biosensor chip using the
standard
amine coupling kit (BlAcore, Uppsala, Sweden). A capture assay was then used
to bind
the antibodies of interest to the anti-human IgG Fc-iznmobilized surface,
aiming for a 300
response unit BlAcore signal. The BlAcore was operated at 25C with a flow rate
of
lOul/min and running buffer containing 10mM Na-HEPES, pH 7.5/ 150mM NaCI/ 3mM
EDTA/ 0.005% Tween 20. After ligand binding, flow rate was increased to
25u1/min and
MN analyte (50nM to 1 nM) was flowed over the surface for 10min such that the
association phase could be monitored. Dissociation then proceeded for 35 min,
followed
by regeneration of the anti-human IgG Fc-immobilized surface at 100ul/n-tin
with 10mM
79

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
phosphoric acid. Sensorgrams were fitted using a 1:1 Langmuir binding model to
calculate rate constants (Table 1, below).
Table 1
Affinity of panning-derived antibodies to the soluble MN determined by surface
plasmon
resonance (Biacore)
1 E4 3EE9 1AA1 3A4 5AA3 3EF2
Affinity 5 4 27 1 1 6
(Kd, nM)
Antibody Binding
3ee9/MMAE was shown to have a kD of 3.6nM for purified MN protein using
Biacore technology, which was the same as the affinity of the unconjugated
antibody 3ee9.
The binding to MN (CA IX) appeared specific, as there was no detectable
binding to 13
other carbonic anhydrases. Binding to mitochondria-associated CA5 was
observed, but
this isozyme would be inaccessible to the antibody in vivo.
By FACS analysis, 3ee9/MMAE was shown to bind to MN-expressing MaTu cells,
but not MN-negative DLD cells (Figure l0a). The MN antibody G250, a humanized
IgGI
antibody, (Wilex) was used as a reference throughout the in vitro studies. It
bound MN
with a Kd of 5.3nM and exhibited a similar binding profile to MN+ and MN- cell
lines as
3ee9/MMAE.
Similar results were obtained for the antibody 1E4 and laal immunoconju gates
(Figures lOb and lOc, respectively), which exhibited binding to MN+ cells, but
not to MN-
cells.
Antibody Internalization
3ee9/MMAE was selectively internalized by MN-expressing cells (PC3mm2), but
not by MN-negative cells (DLD1), as measured by Cellomics (Figure 1 la).
Similarly, the

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
1E4 immunoconjugate was found to be internalized by MN+ cells (PC3mm2), but
not by
MN- (DLD 1) cells (Figure l lb).
Immunoprecipitation
To further explore specificity, various candidate antibodies including the
antibody
portion of 3ee9/MMAE-(3ee9) and the reference antibody G250 were incubated
with cell
lysates of MN+ (Pc3mm2) and MN- (DLD-1) cell lines that had been selectively
labeled
with biotin. Complexes between the antibodies and cellular proteins were
immuno-
precipitated and the co-precipitated antigens visualized using immunoblots
developed with
enzyme-linked streptavidin. Both G250 and the 3ee9 antibody of 3ee9/MMAE
selectively
bound and co-immunoprecipitated with a single band of the same size as MN from
MN+
cells (Figure 12). Less specific antibodies such as 3ef2, 5aa3 and 5A6 co-
immunoprecipitated several other proteins in addition to MN.
Cytotoxicity
In vitro cytotoxicity assays were carried out. 3ee9/MMAE was found to be
highly
cytotoxic to MN-expressing PC3mm2 cells (EC50 = 50 nM), but not to MN-negative
MIAPaca2 cells (Figure 13a). Less than 10% killing of MN-negative cells was
seen even
at doses as high as luM. The lA4 immunoconjugate was also found to be
selectively
cytotoxic for MN+ cells (PC3mm2 and MaTu), but not for MN- cells (MIAPaca2 and
DLDl) in the in vitro cytotoxicity assays (Figure 13b). Additionally, the laal
imrnunoconjugate was found to be selectively cytotoxic for MN+ cells (PC3mm2),
but not
for MN- cells(MIAPaca2) in the in vitro cytotoxicity assays (Figure 13c).
The cytotoxic drug MMAE delivered by 3ee9/MMAE is a tubulin inhibitor that
prevents spindle formation during cell mitosis resulting in G2/M arrest. The
effect of
treatment of MN-expressing cells (Pc3mm2) and MN non-expressing cells (H460)
with
3ee9/MMAE is shown in Figure 14. The tubulin is stained with florescence
labeled
81

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
antibody. Untreated cells showed normal spindle formation, while the treated
MN
expressing cells showed fragmented fibers resulting from tubulin binding with
MMAE and
prevention of normal spindle formation. No activity was seen in MN non-
expressing cells.
These studies confirmed that the antibody drug conjugate 3ee9/MMAE kills cell
through
targeted tubulin disruption.
Example 12. In Vivo Activity of Immunocon.iugate
In vivo pharnzacology of 3ee9/MMAE Against Subcutaneously Implanted Human
Xenograft Cancer Models
3ee9/MMAE exhibits significant and consistent anti-tumor effects on the growth
of
multiple human xenograft tumor models in athymic mice when administered via
the
intravenous route in an intermittent schedule. The in vivo anti-tumor effect
of
3ee9/MMAE was examined in 6 different human xenograft tumor models. These
models
were established through subcutaneous implantation of human tumor cells into
female
athymic NCr (nulnu) mice (Taconic, NY). The human tumor xenograft models
evaluated
included the MaTu human mammary carcinoma model, the HT-29 and Colo-205 human
colo-rectal carcinoma (CRC) models, PC3MM2 prostate carcinoma model, the NCT-
15
multi-drug resistant (P-gp) CRC model, and the MiaPaCa2 human pancreatic
model. PBS
was used as a vehicle, and dosing solutions were prepared fresh daily. The
dosing volume
was 0.1 mlJlOg (10 mL/kg).
The length and width of each tumor were measured using an electronic caliper 2-
3
times per week, and tumor weights (mg) were calculated based on the formula of
[length
(mm) x width (mrn)2]/2. All data, including daily observations, obtained
throughout the
course of the study were documented in Anti-tumor Data Acquisition System
(ADAS).
The maximum tolerated dose (MTD) is defined as the highest dose that does not
produce
greater than 20% lethality and/or 20% net body weight loss. Tumor growth
inhibition
82

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
(TGI) was calculated as (1-T/C)x100, where T = final tumor weights from a
treated group
after the last dose, and C = final tumor weights from the control group after
the last dose.
In addition, anti-tumor efficacy was measured as the incidence of responses
(or responders
or regressions) defined as tumors with S 50% of their initial size. A minimum
duration of
7 days is required for a response to be considered durable.
Efficacy of 3ee9/MMAE in MaTu Xenograft Model
Human mammary xenograft, MaTu cells were maintained as adherent cultures in
RPMI supplemented with 10% FBS. NCr nude mice (8-12 weeks of age) were
inoculated
subcutaneously in the right flank with 5x 106 cells in 0.1 mL of 80%
matrigell20% HBSS.
When tumors reached an average size of -180 mg (7 days), treatment was
initiated. BAY
79-4620 (3ee9-IC) was administered i.v. once every four days (Q4Dx3) at a dose
of 1, 3
and 10 mg/kg. Control mice were treated with PBS or an un-conjugated
monoclonal
antibody at a dose of 10 mg/kg.
Daily examinations into the health status of each animal were conducted. Each
experimental group consisted of 10 mice, and the dosing volume was 0.1 mU10 g
body
weight. The length and width of each tumor was measured by using an electronic
caliper
2-3 times per week, and tumor weights (mg) were calculated based on the
formula of
[length (mm) x width (mm)2]/2. All data, including daily observations,
obtained
throughout the course of the study were documented. Tumor growth inhibition
(TGI) was
calculated as 1-T/Cx 100, where T= final tumor weights from a treated group,
and C
final tumor weights from the control group.
3ee9/MMAE was well-tolerated at all doses examined with all the treated
animals
exhibiting no significant weight loss. Representative efficacy of 3ee9/MMAE in
the
MaTu tumor model is illustrated in Figure 15. Tumors from both the untreated
and
vehicle-treated control groups grew progressively in all animals. The mean
doubling time
83

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
for animals in control and vehicle groups were 11.2 days. At the end of
dosing,
3ee9/MMAE showed robust anti-tumor efficacy at all doses examined. More
specifically,
BAY 79-4620 (3ee9-IC) yielded 67, 72 and 78% TGI at 1, 3 and 10mg/kg,
respectively.
In comparison, the unconjugated 3ee9 mAb had no significant effect in
inhibiting the
growth of this mammary xenograft -tumor. -
Following the completion of the pre-determined dosing regimen (Q4Dx3), the
effect of 3ee91MMAE on tumor growth delay and regression was determined. As
shown
in Figure 15, treatment of 3ee9/MMAE resulted in significant tumor growth
delay and
regression. At the lowest dose examined (lmg/kg), following cessation of
treatment,
tumors remained stable for -2 weeks and began to grow back thereafter.
Overall, 30% of
the tumors were responsive to this treatment of lmg/kg. In comparison, 100% of
the
animals responded when challenged with 3 and 10mg/kg. Of note, even after -3
months
following the cessation of treatment, only 3 and 1 tumors showed signs of re-
growth in the
3 and 10 mg/kg groups, respectively, showing that 70 and 90% of the animals
remained
tumor-free even after -90 days after the cessation of treatment.
The monoclonal IgGI 1E4, when conjugated to a cytotoxic drug, produced a
significant anti-tumor effect against the human mammary xenograft, MaTu, when
dosed at
30 mg/kg immunoconjugate, whereas the unconjugated antibody had no effect
(Figure 16).
Further experiments using the same protocol, but substituting conjugated forms
of
antibody laal (Figure 17), 3ee9 and 5aa3, gave similar results. Additionally,
when using
the conjugated form of 3ee9, anti-tumor effects were seen in other human
xenograft
models derived from various histological type including, HT-29 and Colo-205
human
colo-rectal cancer xenograft models, and PC3-mm2, a human prostate xenograft
model.
These data indicate that the therapeutic utility of antibodies directed
against MN protein as
vehicles for cytotoxic drug delivery to tumors.
84

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Example 13. Therapeutic Index Determination of 3ee9/MMAE in MaTu Xenograft
Model
The maximum tolerated dose (MTD), minimum efficacious dose (MED), and
therapeutic index (TI) of 3ee9/MMAE was determined using MaTu xenograft tumor-
.5 bearing mice.- The TI is defined as the ratio of the MTD divided by MED.
3ee9/MMAE
was administered intravenously once every 4th day for a total of 3 injections
(Q4Dx3).
3ee9/MMAE was administered at dose levels of 0.625, 1.25, 2.5, 5.0, 10, 30 and
60mg/kg.
Control mice were treated with PBS or an unconjugated monoclonal antibody at a
dose of
60 mg/kg. 3ee9/MMAE dosed at 60mg/kg appeared to be the MTD, as there was 10%
lethality and -20% body weight loss in response to this treatment. All other
treatment
groups were well tolerated.
The anti-tumor activity of 3ee9/MMAE is presented in Figure 18. At the end of
dosing, 3ee9IMMAE at a dose of 0.625 and 1.25mg/kg resulted in 62 and 81%
inhibition,
respectively. Doses of 2.5, 5, 10, 30 and 60mg/kg resulted in a greater
inhibition of tumor
growth (-90%) with majority of tumors starting to show regressions. Based on
data
observed, the MED of 3ee9/IVIMAE was 0.625mg/kg. Following the completion of
the
pre-determined dosing regimen (Q4Dx3), the effect of 3ee9/MMAE on tumor growth
delay and regression was determined. No tumor regression was seen for the
0.625mg/kg
dose. In contrast, 80% of the animals exhibited tumor regressions in response
to
1.25mg/kg. Moreover, at doses of 2.5mg/kg and higher, 100% of the animals
responded to
treatment. The therapeutic index of 3ee9/MMAE was determined to be -96.
Example 14. Efficacy of 3ee9/MMAE in HT-29 Xenograft Model
Human CRC xenograft, HT-29 cells were inoculated subcutaneously in the right
flank with 5x 106 cells in 0.1 mL of HBSS. When tumors reached an average size
of - 120
mg (5 days), treatment was initiated. 3ee9/MMAE was adn-unistered i.v. once
every four

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
days (Q4Dx3) at a dose of 0.625, 1.25, 2.5, 5.0, 10 mg/kg. Control mice were
treated with
PBS or an un-conjugated monoclonal antibody at a dose of 10 mg/kg. In
addition, MMAE
was assessed as a free drug at a dose of 0.1, 0.2 and lmg/kg. The 0.1 and 0.2
mg/Kg
doses of MMAE represent equivalent amounts of this drug to those present on 5
and 10
mg/Kg 3ee9/MMAE respectively.
3ee9/MMAE was well tolerated at all doses examined with all treated animals
exhibiting no significant weight loss. Similarly, lower doses of MMAE, i.e.,
0.1 and
0.2mg/kg, were also well tolerated with non-significant, minimal weight loss.
However, at
the top dose of lmg/kg, 50% lethality was observed with the remaining animals
exhibiting
severe weight loss and thus was considered toxic.
Representative efficacy of 3ee9/MMAE and free MMAE in the HT-29 tumor
model is illustrated in Figure 19. Tumors from both the untreated and vehicle-
treated
control groups grew progressively in all animals. The mean doubling time for
animals in
control and vehicle groups were 6.4 days. At the end of dosing, 3ee9/MMAE
showed
robust anti-tumor efficacy at all doses examined. More specifically, 3ee9/MMAE
at doses
of 0.625, 1.25, 2.5, 5 and 10 mg/kg yielded 54, 72, 97, 100 and 100% TGI,
respectively.
In comparison, free MMAE of 0.2mg/kg resulted in significant TGI of 60%,
whereas
0. lmg/kg had no significant effect in inhibiting the growth of this xenograft
model. In
terms of tumor responses, 3ee9/MMAE dosed at 1.25mg/kg resulted in 20% of the
animals
showing regressions. At higher doses, the tumor responses were much greater,
with 2.5
mg/kg showing 90% regressions and 5 and 10mg/kg inducing 100% responses. In
contrast, free MMAE did not induce any tumor responses as defined above.
Anti-tumor effect of 3ee9/MMAE was also assessed in varying schedules, i.e.,
once a week dosing for total of two doses (Q7Dx2, Figure 20) and a single dose
at the time
of staging (Q1Dx1, Figure 21). In both schedules, 3ee9/MMAE was dosed at
0.625, 1.25,
86

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
2.5, 5 and 10 mg/kg. Regardless of schedule, 3ee9/MMAE was highly effective in
inhibiting the growth in this CRC xenograft model.
Table 2, below, summarizes anti-tumor efficacy of 3ee9/MMAE. The anti-tumor
efficacy was very similar to that which was observed for 3ee9/MMAE in the
Q4Dx3
schedule, indicating that in the schedules examined, the anti-tumor efficacy
of
3ee9/MMAE appears to be schedule-independent.
Table 2
Anti-tumor efficacy of 3ee9/MMAE
Dose Treatment % Inhibition Percent
Compound (1-T/D)*100
(mg/kg) Schedule (Day 18) Regressions
Control N/A N/A N/A 0
PBS 0 Q4D x 3 -17 0
3ee9 Ab 10 Q4D x 3 -11 0
3EE9IC 0.625 Q4D x 3 54 0
3EE9IC 1.25 Q4D x 3 72 2
3EE9IC 2.5 Q4D x 3 97 9
3EE91C 5 Q4D x 3 100 10
3EE9IC 10 Q4D x 3 100 10
3EE9IC 0.625 Q7D x 2 48 0
3EE91C 1.25 Q7D x 2 71 0
3EE9IC 2.5 Q7D x 2 92 4
3EE9IC 5 Q7D x 2 96 7
3EB9IC 10 Q7D x 2 100 10
3EE9IC 0.625 Q1D x 1 42 0
3EE9IC 1.25 Q1D x 1 48 0
3EE9IC 2.5 Q1D x 1 93 5
3EE9IC 5 Q1D x 1 98 8
3EE9IC 10 Q 1 D x 1 100 10
MMAE 1 Q4D x 3 Toxic N/A
MMAE 0.1 Q4D x 3 27 0
MMAE 0.2 Q4D x 3 60 0
Exampie 15. Efficacy of 3ee9/MMAE in PC3mm2 and Colo-205 XenoLyraft Models
87

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
The anti-tumor activity of 3ee9/MMAE was next evaluated against human prostate
(PC-3mm2) and CRC (Colo-205) tumor xenografts. Female NCr nu/nu mice were
implanted subcutaneously (s.c.) either with 5 x 106 PC3mm2 or Colo-205 cells.
Treatment
was initiated when tumors were of an average size of approximately 125-150mg.
The
=5 general health of mice was monitored and recorded daily. Tumordimensions
and body
weights were recorded twice a week starting with the first day of treatment.
In the PC3mrn2 and Colo-205 studies, 3ee9/MMAE was administered
intravenously (i.v.) once every 4th day for a total of 3 injections (Q4Dx3) at
doses of 1, 3,
and 30mg/kg and 1.25, 2.5, 5 and 10mg/kg, respectively. As in other studies,
10 3ee9/MMAE was well tolerated at all doses evaluated. 3ee9/MMAE inhibited
the growth
of established PC3MM2 prostate tumors when administered at dose levels of 10
and
30mg/kg, with 100% of the animals showing responses (Figure 22). At the lower
doses of
1 and 3 mg/kg, only moderate effects were seen, as 45 to 50% TGI was observed
at the
end of dosing. 20% of the animals did, however, show responses in the 3mg/kg
group.
Similarly in the Colo-205 CRC xenograft model, 3ee9/MMAE was highly
effective in inhibiting tumor growth (Figure 23). At the end of dosing, >90%
TGI was
observed for 5 and 10 mg/kg doses and >70% TGI was seen in response to
2.5mg/kg dose.
The lowest dose, Img/kg, of 3ee9/MMAE was relatively ineffective against this
CRC
model. In terms of regressions, 3ee9/MMAE dosed at 5 and 10mg/kg resulted
in,40 and
80% responses, respectively.
Example 16. Efficacy of 3ee9/MMAE in HCT-15 Xenograft Model
To examine the effect of 3ee9/MMAE in a multi-drug resistance model, female
NCr nulnu mice were implanted subcutaneously (s.c.) with 5 x 106 HCT-15 cells.
HCT-15
is a multi-drug resistant (MDR) line that over-expresses P-glycoprotein (P-
gp). As such,
this cell line exhibits a multi-drug resistance phenotype and is resistant to
Taxol ,
88

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
doxorubicin and etoposide. 3ee9lMMAE, Taxol, and Gemcitabine were administered
when mice had established tumors with mean weight of - 150 mg. Gemcitabine, a
pyrimidine analog that is not a P-gp substrate was used as a positive control.
As expected,
Gemcitabine (120 mg/kg, i.p., QDx10) was highly active in this MDR model
(Figure 24).
-5 In contrast, 3ee9/MMAE (MMAE being a known substrate of P-gp) and= Taxol
failed to
produce any significant anti-tumor effect.
Error!Example 17. Efficacy of 3ee9/MMAE in huMN-MIAPaca2 and MIAPaca2
Xenograft Models
To better understand the relationship between MN expression and antitumor
efficacy, xenograft efficacy studies were conducted using a low MN-expressing
pancreatic
model, MIAPaCa2 and high MN-expressing huMN-MIAPaCa2 model. This latter line
was derived by engineering the MIAPaCa2 line to stably express MN. 3ee9/MMAE
had
minimal effect in inhibiting the growth of this low MN-expressing MIAPaCa2
line (Figure
25). 3ee9/MMAE dosed at 10mg/kg only produced 50% TGI. In contrast,
significant
anti-tumor activity was seen for 3ee9/MMAE against huMN-MIAPaCa2 model, where,
at
the end of dosing, doses of 2.5, 5 and 10mg/kg yielded TGI of 63, 82 and 94%,
respectively. Moreover, the 10mg/kg dose induced 100% responses, indicating
that there
is a great shift in sensitivity by over-expressing MN levels in tumors.
Example 18. Combination with XelodaO in Colo-205 Xenograft Model
The feasibility of using 3ee9/MMAE in conjunction with other cancer
chemotherapeutic agents was investigated. Xeloda is used as first line
treatment of
patients with metastatic colorectal carcinoma. The activity and tolerability
of combined
therapy using 3ee9/MMAE and Xeloda was evaluated. 3ee9/1VIMAE was
administered
i.v. on a Q4Dx3 schedule and at dose levels of 1.25, 2.5 and 10 mg/kg. Xeloda
was
administered orally once daily for 9 days at dose levels of 250 and 500 mg/kg.
89

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
3ee9/MMAE administered alone at 2.5 and 10 mg/kg resulted in robust tumor
growth
inhibition (TGI of 73 and 96%, respectively) (Figure 26a). The 1.25mg/kg group
of
3ee984MAE, however, was relatively ineffective, resulting in only 31% TGI.
Xeloda
administered alone at dose levels of 250 and 500 mg/kg also produced robust
TGI of 78
and 86%, respectively.
In terms of responses, 10mg/kg group of 3ee9/MMAE induced 80% response rate,
whereas zero percent of the animals in both dose groups of Xeloda showed any
regressions. All combination treatment groups were well tolerated and resulted
in
significant inhibition of tumor growth as well response rates. Quantitative
data detailing
the TGI and tumor responses by combining these two agents are shown in Figures
26a and
26b, as well as Table 3, below. In the doses examined, the anti-tumor activity
of
3ee9/MMAE in combination with Xeloda was far superior to that of either
3ee9/MMAE
or Xeloda administered as single agents. Finally, in regards to tolerability,
administration
of these therapeutics was well tolerated with no adverse reactions.
Table 3
Anti-tumor activity of BAY 79-4620 (3ee9-IC) in combination with Xeloda'O
Treatment Schedule Dose % Jo Weight % Re-
mg/kg/i [nhibition Loss (d 17) gression
nj (T/C) (d s
17)

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Untreated - - - 0.3 0
79-4620 (3ee9 cj) Q4DX3;IV 1.25 30.6 0.5 0
79-4620 (3ee9 cj) Q4DX3;IV 2.5 73 0.1 0
79-4620 (3ee9 cj) Q4DX3;IV 10 96.2 0.9 80
Xeloda Q1DX9;PO 250 - 78= ---0.5 0
Xeloda Q1DX9;PO 500 86.1 -5.6 0
Xeloda (250
mg/kg)
Q4DX3;IV/Q1 1.25 92.1 -2 50
79-4620 DX9;PO
(3ee9cj)+Xeloda
79-4620 (3ee9 Q4DX3;IV/Q1 2.5 91.4 -0.9 60
cj)+Xeloda DX9;PO
79-4620 (3ee9 Q4DX3;IV/Ql 10 97.5 -0_4 100
cj)+Xeloda DX9;PO
Xeloda (500
mglkg)
79-4620 (3ee9 Q4DX3;IV/Q1 1.25 94.2 -0.3 70
cj)+Xeloda DX9;PO
79-4620 (3ee9 Q4DX3;IV/Q1 2.5 95.7 -3.2 90
cj)+Xeloda DX9;PO
79-4620 (3ee9 Q4DX3;IV/Q1 10 97.3 -1.4 100
cj)+Xeloda DX9;PO
Example 19. In vivo Distribution / Tumor Localization of 3ee9 mAb AEainst
Bilaterally Implanted Human Xenograft Cancer Model
To determine the in vivo distribution and tumor localization of the mAb
component
s of 3ee9/MMAE, non-invasive in vivo imaging studies were conducted using the
CRI
MaestroTM in mice which received bilateral implantation of tumors exhibiting
both high
and low MN expression. The MaestroTM in-vivo imaging system (CRI, Woburn, MA)
with multi-spectral acquisition and analysis is designed to eliminate auto-
fluorescence
91

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
background. Mabs 3ee9 of BAY 79-4620, M75 (mouse monoclonal recognizing MN)
and
control human IgG, were conjugated with Alexa Fluor 750 (Invitrogen Cat# A2001
1)
according to manufacturer's instructions. The ratio of Protein/AF750(mol/mol)
was 1/5.9
for 3ee9, 8.9 for human IgG, and 6.0 for M75.
.5 Twelve days before= the injection of each fluorescence-labeled antibody,
Harlan
Balb/C nudes were implanted with MIAPaCa-2 (low MN-expressing; 7.5x 106 cells
in 50%
Matrigel) subcutaneously in the right flank, and huMN-MIAPaCa2 (MN transfected
stable
cell line; 5x106 cells in 50% Matrigel) subcutaneously in the left flank. 4 g
of conjugated
antibody was injected on Day 0 (12 days post implant) to each animal. Imaging
was done
on Day 4, Day 5 and Day 10. Data were collected using animals that were
anaesthetized
(100mg/kg ketamine /10mg/kg xylazine, i.p.), and placed inside the imaging
system.
Multi-spectral image cubes (series of images) were acquired with images spaced
every 10
nm throughout the spectral range of 680 to 950nm, which covered the Near
Infrared (NIR)
range.
Each image was exposed for 5 seconds. False-colored images were synthesized
from the spectral cube using Maestro software and scaled to visible brightness
using
ImagePro Plus 6Ø Scaling was identical to all images, with the brightest
image slightly
below saturation level. Most fluorescently labeled antibodies tend to localize
to the liver
and the bladder. After 4 to 5 days, most of signal then decreases from the
liver and the
bladder and stabilizes in the tumor. Images taken on Day 5 yielded the highest
signal to
background ratio (Figure 27.) After five days, there was very little signal
generated from
the hIgG-injected animals, indicating that hIgG failed to localized into tumor
tissue. In
contrast, both M75 and 3ee9 localized specifically to high MN-expressing (huMN-
MIAPaCa2) tumors. No localization was seen in low MN-expressing MIAPaCa2
tumors
in the same animals.
92

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Examnle 20. In vivo Mechanism of Action
To explore the in vivo mechanism of action of 3ee9/MMAE, tumors were
implanted subcutaneously at right flank of the mice with 5x106 HT-29 cells.
Eight days
later, athymic mice bearing HT-29 tumors were treated with vehicle or with
3ee9/MMAE
.5 at 1.25 and 5 mg/kg (Q1Dxl). -Tumors were collected and fixed in formalin 4
hours and 1,
3 and 5 hours following administration of 3ee9/MMAE. Samples were then
embedded in
paraffin, sectioned at 5 m, de-paraffinized, and stained using standard
protocol for
fluorescent immunohistochemistry of human tissues using mouse antibodies: Anti-
ct/0-
tubulin, Anti-phospho-Histone H3, and DNA. The slides were then observed under
a
fluorescent microscope, and representative images were taken through three
separate color
channels.
3ee9/MMAE had little effect in affecting cell mechanism in the 4hr samples.
However, by day 1, increased numbers of cells were in G2/M arrest, and multi-
polar
spindle was clearly seen. In addition, decreased level of tubulin staining
could be
observed. These effects were then highly amplified in the day 3 and day 5
samples. In
fact, almost all cells in the 5 mg/kg dose group of 3ee9/MMAE on day 5 were
severely
affected by treatment (Figure 28). These data clearly indicate that the
3ee9/MMAE
affected the growth of cancer cells by tubulin inhibition, leading to G2/M
arrest and
apoptosis.
Table 4, below, summarizes the behavior profile of 3ee9/MMAE.
Table 4
3ee9/MMAE profile
Assay Result for 3ee9/MMAE
Affinit (Kd) 3.6 nM
Cell binding (FACS): MN+/MN- +++/-
Internalization: MN+/MN- +++/-
Cytotoxicit : MN+/MN- (ec50) 50 nM/>1gM
Inununo recipitation specific
93

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Assay Result for 3ee9/MMAE
IHC specific
In vivo distribution normal
In vivo activity: MED 1 mg/kg
In vivo activity: MTD 60 mg/k
Example 21. Expression vector construction, transfection, expression and
purification of anti-MN IgGs based -from 3ee9 light and heavy CDR-variable
regions
using a stable CHO cell expression system
Construction of expression vector 3ee9 ,t pCMVuc
The kappa and heavy CDR variable regions (SEQ ID NOS: 126 and 125,
respectively) from vector 3ee9pMORPHx9 (obtained in accordance with Examples 1-
3)
were inserted into vector H+LpattB (ML Laboratories) as follows. Approximately
350
base pair EcoRV - BsiWI restriction enzyme fragment from 3ee9pMORPHx9
(prepared in
accordance with the previous Examples, e.g. Examples 1-3) was inserted into
the EcoRV -
BsiWI restriction enzyme sites of H+LpattB to generate vector 3ee9kappapAttB.
Next,
the approximately 350 base pair MfeI-BlipI restriction enzyme fragment from
3ee9pMORPHx9 was inserted into the EcoRI - B1pI restriction enzyme sites of
3ee9kappapAttB to generate 3ee9H+LpattB. The 3ee9 heavy and light coding
sequences
(SEQ ID NOS: 126 and 125, respectively) were then recombined with pDONR221
(Invitrogen Cat.# 12536-017) using Gateway BP Clonase II enyme mix (Invitrogen
Cat.#
11789-100) to generate vector 3ee9H+LpENTR. Vector 3ee9H+LpCMVucoE8 was
generated by recombination between 3ee9H+LpENTR and pCMV_UCOE8_DEST using
Gateway LR Clonase II enyme mix (Invitrogen Cat.# 11791-100)_
The construction of pCMV_UCOE8_DEST was as follows. Gateway vector
conversion cassette (Invitrogen; cat# 11828-029) was inserted into the Smal
site of
pCET906 to generate vector pCET906_gw. Vector pCET906 was obtained from ML
Laboratories and described fully in Williams et. al., BMC Biotechnology,
(2005), 5:17,
94

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
which is incorporated herein by reference. The approximately 2900 base pair
Agel
restriction enzyme fragment from pCET906_gw was then cloned into the Agel site
of
pCET1015 to generate pCMV_UCOES_DEST. Vector pCET1015 was also obtained
from ML Laboratories and which is described in Williams et al., supra. The
vector
= 5 pCMV_UCOE8_DEST was propagated in an E. coli strain resistant to the ecdB
toxin gene
(e.g. such as those marketed as " One Shot ccdB Survival T1 cells" from
Invitrogen (cat#:
C7510-03)). The complete nucleotide sequence of the insert of 3ee9H+
.pCMVucoF8 is
shown in Figure 29, i.e. the complete nucleotide sequence encoding an human
IgG anti-
MN antibodYcomprising the kappa and heavy CDR variable regions of SEQ ID NOS:
126
and 125, respectively, obtained from vector 3ee9pMORPHx9.
Isolation of cell clone 3ee9.25
To transfect CHO-S cells with vector 3ee9H+L.PCMVUCOE8, 60 pg of DNA was
diluted in 2 mLs of CD CHO (invitrogen#10743-029) complete with 8mN1 L-
glutamine
and HT(invitrogen) and lacking PenStrep. Next 2 mis of CD CHO complete media
was
added to a 250 mis erlenmeyer flask. Then, 150 pL of DMRIE-C (Invitrogen
cat#10459-
014) was added to the flask and incubated at RT for 10 min. The diluted DNA
was then
mixed with the diluted DMRIE-C, flushed with 5% CO2 and incubated at RT for 30
min.
During the incubation, 4 mis of CHO-S cells at 5x106 c/mL (2x107 total cells)
were
prepared in CD CHO complete (no P/S). After the incubation, the cells were
added to the
flask with DNA-DMRIE-C complex, and then gently swirled to mix. The flask was
then
flushed with 5% CO2 and shaken at 125 rpm at 37C for 4 hrs. Next, 32 mis of CD
CHO
complete media with no Pen Strep was added to the flask, flushed with 5% CO2i
and then
returned to shaker (37 C) overnight. After 24hrs, cells were counted, spun
down, and
resuspended in CD CHO complete plus 5.5 mL/L PenStrep with 20% conditioned
media
and antibiotic (12.5 g/ml puromycin) at appropriate cell density for plating.
Cells were

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
plated at 100, 300, 900, and 2700 cells/well in 96 well plates. The plates
were then
incubated in a 5% COZ incubator for approximately 3 weeks. Care was taken not
to
disturb the plates.
Individual clones were expanded from plates having wells containing single
=5 colonies in less than 20%=of the wells=on the plate. The cells were
expanded to non-tissue
treated 24 well plates with 1 ml selective media per well. Cells were
incubated for about 1
week during which 500 pL of fresh media was added as needed. Antibody
expression was
tested at a single dilution to eliminate clones that do not express. Clones
were tested by
ELISA in duplicate using a 1:5 dilution (5uL supernatant in 95uL TBS/tween).
Clones
were eliminated that gave an OD (optical density) below 0.1. Positive clones
were
expanded into non-tissue treated 6 well plates. Next 5 mLs of cells seeded at
4x 104
ce11s/mL. After 4 days of incubation protein expression was determined by
ELISA.
Clones were tested using 1:50, 1:100, 1:200, and 1:400 dilutions. Clone
3ee9.25 exhibited
the highest secreted antibody concentration. The 3ee9.25 cells were transfered
into a 125
mL erlenmeyer shaker flask seeding at 2x 105 cells/mL to adapt cells to
suspension growth.
Expression of the anti-MN IgGs
Transfected CHO-S cells were seeded into a Wave 10 Liter Bioreactor (Wave
Biotech, 300 Franklin Square Drive, Somerset, New Jersey, 08873, USA) at
500,000
cells/ml. Cells were cultured for seven days in 50% CD-CHO Media (Invitrogen
10743),
50% Sigma CHO media Number 5 (Sigma 0363), 1% FCS, lx HT supplement
(Invitrogen
11067-030), Penicillin/Streptomycin (Invitrogen 15140-122), 8 mM L-Glutamine
(Invitrogen 25030-08 1), and 12.5 g/m1 Puromycin (BD 631305) with the
bioreactor
rocking at 25 rpm under a 5% CO2 atmosphere at 37 C. At the end of the
fermentation
period, spent culture media was harvested by centrifugation and sterile
filtered (0.211M)
prior to IgG purification.
96

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
Purification of the anti-MN IgGs.
Typically 10 to 20 L of IgG-containing cell culture media were concentrated 2
to 5
fold using Prep-Scale TFF-2 30 kD Cartridge (Millipore). 1M Tris-Cl buffer, pH
7.5 was
added to the concentrated media to the final concentration of 50 mM. 5.0 M
NaCI was
==5 then added to a final concentration of 150 mM. The concentrated media= was
typically
loaded onto a 30 mL Protein Sepharose column equilibrated with PBS, pH 7.4.
The
column was washed with PBS pH 7.4 containing 0.1% Tween 20 and 1mM EDTA. The
column was then washed with PBS and eluted with 100 mM glycine buffer pH 3Ø
Upon
collection the fractions were neutralized to pH 7.5 with 1M Tris-Cl pH 7.8.
The purified
IgGs were transferred into PBS by dialysis and sterilized by filtration
(0.2pM). Final
purified antibody preparations were adjusted to between 1 and 5 mg/mL,
exhibited a
purity of > 95% as determined by SDS PAGE gels stained with coomassie blue,
whereupon the protein migrated as two bands corresponding to the heavy chains
of Mr =
50 kDa and the light chains of Mr 25 kDa, had endotoxin levels of less then 1
EU/mg of
protein, and less than 10% protein aggregate as determined by SEC HPLC.
Preparations
were also subjected to functional Q.C. for MN antigen binding by surface
plasmon
resonance, binding to MN-expressing cells by FACS, and internalization into
the MN-
expressing cells as determined by automated imaging (Cellomics). A 20L
combined
fermentation typically yielded 1 gram of purified protein with an overall
recovery of 50 to
75 %. N-terminal sequencing of 255 pmol of the final preparation of purified
full length
3ee9 antibody using an ABI Procise 494 HT sequencer yielded 368 pmol of the
following
expected sequence for the mature kappa light chain of: DIQMTQSPSSLSASVGDRVTIT-
RASQDINNYLSWYQQKP-. This is the same as the N-terminal sequence of
corresponding to the mature Vkappal light chain of mAb 3ee9 (SEQ ID NO: 146).
The
97

CA 02633038 2008-06-11
WO 2007/070538 PCT/US2006/047445
heavy chain was not detected by Edman sequencing showing that its sequence was
blocked.
.5 Having thus described in detail preferred embodiments of the present
invention, it
is to be understood that the invention defined by the above paragraphs is not
to be limited
to particular details set forth in the above description as many apparent
variations thereof
are possible without departing from the spirit or scope of the present
invention.
98

Representative Drawing

Sorry, the representative drawing for patent document number 2633038 was not found.

Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Application Not Reinstated by Deadline 2016-08-26
Inactive: Dead - Final fee not paid 2016-08-26
Deemed Abandoned - Conditions for Grant Determined Not Compliant 2015-08-26
Notice of Allowance is Issued 2015-02-26
Letter Sent 2015-02-26
Notice of Allowance is Issued 2015-02-26
Inactive: Approved for allowance (AFA) 2015-01-29
Inactive: QS passed 2015-01-29
Amendment Received - Voluntary Amendment 2014-06-19
Inactive: S.30(2) Rules - Examiner requisition 2013-12-19
Inactive: Report - No QC 2013-12-12
Letter Sent 2013-07-17
Amendment Received - Voluntary Amendment 2013-06-28
Reinstatement Requirements Deemed Compliant for All Abandonment Reasons 2013-06-28
Reinstatement Request Received 2013-06-28
Inactive: Abandoned - No reply to s.30(2) Rules requisition 2012-07-03
Inactive: S.30(2) Rules - Examiner requisition 2011-12-30
Amendment Received - Voluntary Amendment 2011-03-22
Inactive: S.30(2) Rules - Examiner requisition 2010-09-22
BSL Verified - No Defects 2009-09-22
Letter Sent 2009-06-04
Letter Sent 2009-06-04
Letter Sent 2009-06-04
Inactive: Single transfer 2009-03-30
Inactive: Cover page published 2008-10-01
Inactive: Sequence listing - Amendment 2008-10-01
Amendment Received - Voluntary Amendment 2008-10-01
Letter Sent 2008-09-30
Inactive: Declaration of entitlement/transfer - PCT 2008-09-29
Inactive: Notice - National entry - No RFE 2008-09-29
All Requirements for Examination Determined Compliant 2008-07-28
Request for Examination Requirements Determined Compliant 2008-07-28
Request for Examination Received 2008-07-28
Inactive: First IPC assigned 2008-07-08
Application Received - PCT 2008-07-07
National Entry Requirements Determined Compliant 2008-06-11
Application Published (Open to Public Inspection) 2007-06-21

Abandonment History

Abandonment Date Reason Reinstatement Date
2015-08-26
2013-06-28

Maintenance Fee

The last payment was received on 2015-11-19

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
BAYER HEALTHCARE LLC
Past Owners on Record
GERALD RANGES
LILA ADNANE
PAMELA TRAIL
PAUL TAMBURINI
SHA HA
TIMOTHY MCCABE
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2014-06-18 100 4,390
Description 2014-06-18 39 942
Description 2008-06-10 98 4,681
Drawings 2008-06-10 49 1,321
Claims 2008-06-10 24 780
Abstract 2008-06-10 1 58
Description 2008-09-30 100 4,715
Description 2008-09-30 39 942
Description 2011-03-21 100 4,402
Claims 2011-03-21 6 210
Description 2011-03-21 39 942
Description 2013-06-27 100 4,393
Description 2013-06-27 39 942
Claims 2013-06-27 5 184
Claims 2014-06-18 4 135
Acknowledgement of Request for Examination 2008-09-29 1 175
Reminder of maintenance fee due 2008-09-28 1 111
Notice of National Entry 2008-09-28 1 193
Courtesy - Certificate of registration (related document(s)) 2009-06-03 1 103
Courtesy - Certificate of registration (related document(s)) 2009-06-03 1 102
Courtesy - Certificate of registration (related document(s)) 2009-06-03 1 102
Courtesy - Abandonment Letter (R30(2)) 2012-09-24 1 164
Notice of Reinstatement 2013-07-16 1 170
Commissioner's Notice - Application Found Allowable 2015-02-25 1 162
Courtesy - Abandonment Letter (NOA) 2015-10-20 1 164
PCT 2008-06-10 9 275
Correspondence 2008-09-28 1 25
PCT 2010-06-21 1 45
PCT 2010-07-19 1 51

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :